Sunteți pe pagina 1din 27

Review

Environmental Exposure to Mercury and Its Toxicopathologic


Implications for Public Health

Paul B. Tchounwou, Wellington K. Ayensu, Nanuli Ninashvili, Dwayne Sutton


Cellomics and Toxicogenomics Research Laboratory, NIH Center for Environmental Health,
School of Science and Technology, Jackson State University, 1400 Lynch Street,
Box 18540, Jackson, Mississippi 39217, USA

Received 23 February 2003; revised 20 March 2003; accepted 20 March 2003

ABSTRACT: Mercury is a toxic and hazardous metal that occurs naturally in the earth’s crust. Natural
phenomena such as erosion and volcanic eruptions, and anthropogenic activities like metal smelting and
industrial production and use may lead to substantial contamination of the environment with mercury.
Through consumption of mercury in food, the populations of many areas, particularly in the developing
world, have been confronted with catastrophic outbreaks of mercury-induced diseases and mortality.
Countries such as Japan, Iraq, Ghana, the Seychelles, and the Faroe Islands have faced such epidemics,
which have unraveled the insidious and debilitating nature of mercury poisoning. Its creeping neurotoxicity
is highly devastating, particularly in the central and peripheral nervous systems of children. Central
nervous system defects and erethism as well as arrythmias, cardiomyopathies, and kidney damage have
been associated with mercury exposure. Necrotizing bronchitis and pneumonitis arising from inhalation of
mercury vapor can result in respiratory failure. Mercury is also considered a potent immunostimulant and
-suppressant, depending on exposure dose and individual susceptibility, producing a number of patho-
logic sequelae including lymphoproliferation, hypergammaglobulinemia, and total systemic hyper- and
hyporeactivities. In this review we discuss the sources of mercury and the potential for human exposure;
its biogeochemical cycling in the environment; its systemic, immunotoxic, genotoxic/carcinogenic, and
teratogenic health effects; and the dietary influences on its toxicity; as well as the important consider-
ations in risk assessment and management of mercury poisoning. © 2003 Wiley Periodicals, Inc. Environ
Toxicol 18: 149 –175, 2003.
Keywords: mercury exposure; systemic toxicity; immunotoxicity; health risk assessment; management

Correspondence to: Paul B. Tchounwou; e-mail: paul.b.tchounwou@


jsums.edu.
Contract grant sponsor: National Institutes of Health, through Biomed-
ical Research Infrastructure Network–Mississippi Functional Genomics
Network, contract grant number: 5P20RR16470-02/USM-GR00978-04.
Contract grant sponsor: U.S. Department of the Army; through the
MACERAC program, contract grant number: DACA-42-02-C-0057.
Published online in Wiley InterScience (www.interscience.wiley.com).
DOI 10.1002/tox.10116

© 2003 Wiley Periodicals, Inc.

149
150 TCHOUNWOU ET AL.

CONTENTS

1. Abstract ........................................................................................................................................................ 149


2. Keywords ..................................................................................................................................................... 149
3. Abbreviations ............................................................................................................................................... 150
4. Introduction .................................................................................................................................................. 151
5. Sources of Mercury and Potential for Human Exposure ........................................................................... 152
6. Biogeochemical Cycling of Mercury .......................................................................................................... 153
7. Production and Uses of Mercury ................................................................................................................ 154
8. Health Effects Associated with Mercury Exposure ................................................................................... 154
8.1. Elemental Mercury (Hg0) .................................................................................................................... 154
8.2. Inorganic Mercury Compounds .......................................................................................................... 155
8.3. Organic Mercury Compounds ............................................................................................................. 156
9. Immunotoxic Effects of Mercury ................................................................................................................ 157
9.1. Mercury-Induced Immune Responses ................................................................................................. 157
9.2. Mercury-Induced Immune Responses and Apoptosis ........................................................................ 160
9.3. Significance of Mercury-Induced Autoimmunity ............................................................................... 161
10. Genotoxic, Carcinogenic, and Teratogenic Effects of Mercury ................................................................ 163
11. Dietary Influences on Mercury Toxicity and Counter Effects on Diet ..................................................... 164
11.1. Minerals ............................................................................................................................................. 165
11.2. Vitamins ............................................................................................................................................. 165
11.3. Combined Nutrient Effects ................................................................................................................ 165
12. Mercury Risk Assessment and Management .............................................................................................. 166
12.1. Minimum Risk Levels ....................................................................................................................... 166
12.2. Diagnosis of Mercury Intoxication ................................................................................................... 167
12.3. Treatment Schedules .......................................................................................................................... 168
13. Conclusions .................................................................................................................................................. 168
14. References .................................................................................................................................................... 169

Abbreviations: Ab—antibody; Ag—antigen; ALS—amyotropic lateral sclerosis; AMG—autometallography;


AnoAbs—antinucleolar autoantibodies; APC—antigen-presenting cells; BBB— blood– brain barrier; CCB—
calcium channel blockers; CNS— central nervous system; DN— double negative; DR— death receptors; EtHg—
ethylmercury; GBM— glomerular basement membrane; H-2—murine histocompatibility complex; HgIA—
mercury-induced autoimmunity; HgICD—mercury-induced cell death; HRBC— horse red blood cells; ICGN—
immune-complex glomerulonephritis; Igs—immunoglobulins; I-Hg—inorganic mercury; mAb—monoclonal
antibody; MeHg—methylmercury; MHC—major histocompatibility complex; mg/kg b.wt—milligram per kilo-
gram of body weight; NMDA—N-methyl-D-aspartate; PCA—polyclonal cell activation; PCD—programmed cell
death (apoptosis); PNS—peripheral nervous system; ppm—parts per million; PWM—pokeweed mitogen; RfD—
reference dose; SLE—systemic lupus erythematosus; snRNP—small nucleolar ribonucleoprotein particles;
ww—wet weight
ENVIRONMENTAL EXPOSURE TO MERCURY 151

INTRODUCTION people exhibited symptoms of mercury poisoning. It was


finally discovered that the organomercurial was being con-
Although mercury has no known beneficial effect in human centrated in marine animals that were part of the staple diet
physiology, it is estimated that an average man weighing 70 of the local population. Since then similar situations have
kg has an amount equivalent to 13 mg of mercury in his arisen in Sweden, in a population living on fish collected
system (Pier, 1975). It is a highly reactive molecule that from water contaminated by an organic mercury compound
produces toxic effects by binding highly to sulfhydryl and to used as a pesticide (D’Itri, 1972); in Iraq in 1974, among
a lesser degree to hydroxyl, carboxyl, and phosphoryl people exposed to mass mercury poisoning from rice treated
groups. It is widely distributed as an environmental and with pesticide (Amin-Zaki et al., 1981); and in Ghana,
industrial pollutant. In humans high mercury levels are where ethylmercury-treated maize was consumed by a local
found in the skin, nails, hair, and kidneys. The earth’s crust community, with 20 of the 144 cases of mercury poisoning
contains 0.5 parts per million of mercury. The prominent ending in fatalities. In China in the 1970s cases of ethyl-
sources of human exposure to mercury nevertheless come mercury poisoning were reported. The exposure pathway
from dental amalgams, pharmaceuticals, cosmetics, and was the same as in Iraq, where farmers consumed ethylmer-
food, primarily contaminated fish (Eley, 1997). cury chloride–treated rice (Goldman and Shannon, 2001).
Three forms of mercury are found in the environment. Mercury damages DNA (Sager et al., 1984), impairs
Each has specific solubility, chemical reaction, and toxicity mitosis, and disrupts neuronal migration (Hammond, 1971).
characteristics (Clarkson, 2002; Goldman and Shannon, It is therefore neurotoxic during prenatal and postnatal
2001). Quicksilver, or elemental mercury, is released via periods. However, the effects of mercury intoxication are
degassing from the earth’s crust and oceans, and the com- very pronounced on the fetus. The net results of mercury
bustion of fossil fuels releases elemental mercury to the intoxication include mental retardation, cerebral palsy, sei-
environment (ATSDR, 1999). Inorganic mercurous (calo- zures, and ultimately death (WHO, 1990). The kidneys
mel) and mercuric salts have been utilized for medicinal among all the organs are highly susceptible to mercury salts.
purposes through the centuries and employed as detonators Nephrotoxicity manifests itself clinically as acute tubular
in explosives, respectively (Klassen, 1990). necrosis and immunologic glomerulonephritis (Hultman et
Microorganisms in streams and rivers are capable of al., 1998; Tominack et al., 2002). Arrhythmias and cardio-
biomethylating mercuric compounds to form organic spe- myopathy have also been associated with mercury toxicity.
cies of mercury, including the methyl, ethyl, and phenyl Cardiomyopathic patients have been found to have very
types that find their way into the industrial environment. high concentrations of mercury in their hair, thousand of
The uses of these forms of mercury as biocides, pesticides, times above those of controls (Frustaci et al., 1999).
and household antiseptics have contributed to the wide Phenylmercury, once used as a fungicide in diaper rinses
distribution of this heavy metal into the environment. and in latex paint, has been reported to cause acrodyna with
Therefore, it is not surprising that despite efforts to control painful extremities in infants (Agocs et al., 1990), peripheral
its use and release into the atmosphere, mercury continually neuropathy, and hypertension with renal tubular dysfunc-
accumulates in waters and rivers. tions (Roman-Franco et al., 1978; Langford and Ferner,
Methylmercury, the most toxic of the organic forms, is 1999). In adults a severe neurogenic pain syndrome requir-
taken up by fish species, providing a route to move high up ing inpatient pain management is a problem that can de-
the food chain to enter the diet and cause concerns about velop into a type of severe motor neuropathy that has the
public health (Tan and Perkin, 2000). The methylmercury signs and symptoms of both axonal degeneration and Güil-
content of fish varies, by species, by size, and by harvest lain-Barré-like illnesses (Schwarz et al., 1996). The litera-
location. Fish caught in Lake St. Clair, Canada, with mer- ture also contains numerous case reports of reversible neu-
cury levels of 0.07– 0.11 ␮g/g in 1935 had mercury con- rologic conditions associated with mercury exposure
centrations of 0.2–7.0 ␮g/g of body weight by 1970. Tuna characterized as amyotropic lateral sclerosis–like illness
has an average mercury concentration of between 0.13 and (ALS; Adams et al., 1983; Schwarz et al., 1996). Despite
0.25 ␮g/g of body weight (Dales et al., 1971; Hammond, such presentations it is difficult to determine the reference
1971). The daily absorption of all forms of mercury from dose (RfD) of mercury, the highest dose that the body can
fish and seafood is estimated to be 2.3 and 0.3 ng per day tolerate over a lifetime without any side effects. The reason
from all other foods and air and water (Goyer, 1996). for this difficulty is partly attributed to how poorly under-
The problems associated with mercury poisoning were stood is the individual susceptibility that is presumably
not recognized until the occurrence in Japan of what is now related to variations in dietary habits among different pop-
popularly known as Minamata disease (Clarkson, 1997). A ulations.
chemical plant in Minimata Bay used an inorganic mercury Certain individuals are genetically susceptible to devel-
compound as a catalyst in a chemical process, unaware that oping spontaneous autoimmune diseases. The etiology and
this process resulted in the catalytic conversion of the chem- pathogenicity of these, mostly systemic, autoimmune states
ical to an organomercurial. Years later a large number of have been difficult to trace. Recent immunological findings
152 TCHOUNWOU ET AL.

confirm that most origins of these idiopathic autoimmune mosomes 1, 4, and 7 (Kono et al., 1994; Morel et al., 1994;
diseases can be traced to the contamination of the biosphere Drake, 1995); these have equivalent syntenies in human loci
with xenobiotic compounds, among which mercury has (Duits et al., 1995; Salmon et al., 1996; Moser et al., 1998)
been strongly implicated. Low levels of mercury exposure that seem to be ethnically distinct (Duits et al., 1995;
(⬍40 ␮g/kg body weight) in adults may be unsafe in pa- Salmon et al., 1996). This finding probably foretells that in
tients who have a predisposition to develop systemic lupus dealing with xenobiotics like mercury, surprises are likely in
erythematosus (SLE). Acute renal tubular lesions and im- genetic MHC and non-MHC SLE susceptibility among dif-
munosuppression follow exposure to large doses, whereas ferent racial groups, and so treatments should be scheduled
chronic administration of smaller doses of mercury leads to accordingly.
the development of SLE (Bariety et al., 1971; Roman- Evidence is gradually emerging from HgIA studies that
Franco et al., 1978; Kasturi et al., 1995). Mercury-induced xenobiotic compounds have a characteristic like mercury
autoimmunity (HgIA) shares the same pathogenicity and that induces autoimmunity in genetically predisposed indi-
clinical manifestations seen in patients suffering from SLE viduals. Because of the public health significance associated
(Hirsch et al., 1982; Dubey et al., 1991; Mathieson et al., with environmental exposure to mercury, developing a
1992); the symptomatology is also the same (Jiang and health advisory/criteria document for this toxic and hazard-
Möller, 1995; Biancone et al., 1996; Kono et al., 1998). ous metal is important. In this research report we provide
The experimental development of HgIA relied not only and discuss relevant information on the sources of mercury
on the amount and duration of the metal exposure, but also and the potential for human exposure; the general health
on the genetic background of the exposed animal (Hultman effects associated with exposure to elemental, inorganic and
et al., 1992; Hultman et al., 1993; Jiang and Möller, 1995; organic forms of mercury; immunotoxic, genotoxic, carci-
Kono et al., 1998; Hanley et al., 2002; Pollard et al., 2002). nogenic, and teratogenic effects of mercury; and dietary
Murine experiments have demonstrated that autoantibody influences on mercury toxicity; as well as important con-
specificities and susceptibility to immune complex genera- siderations in mercury risk assessment and management.
tion depend on the H-2 haplotype of the mouse. Evidence Such information is critical in developing efficient and
has also pointed to the involvement of non-MHC genes in effective strategies for dealing with the health hazard asso-
HgIA susceptibility. The major histocompatibility complex ciated with mercury exposure.
(MHC) genes, which in humans are on chromosome 6, are
represented in mice by the H-2 region, found on chromo-
some 17. Immunological studies of various haplotype
SOURCES OF MERCURY AND POTENTIAL
strains of mice have revealed significant differences in
FOR HUMAN EXPOSURE
susceptibilities to mercury. For example, BALB/c mice of
the H-2d haplotype are highly susceptible to HgIA lympho-
Potential Sources of Mercury
proliferation, although they fail to produce immune-com-
plex glomerulonephritis (ICGN; Jiang and Möller, 1995). Mercury occurs naturally. However, natural phenomena
B10.D2 mice are capable of lymphoproliferation and de- such as the erosion of mineral deposits and volcanoes and
velop less severe ICGN than BALB/c mice, whereas the human activities such as metal smelting, coal production,
DBA/2 strain of mice, also of the H-2d haplotype, is resis- chemical synthesis and use, and waste disposal may lead to
tant to both lymphoproliferation and ICGN (Hultman et al., substantial contamination of the environment. Mercury is
1992; Jiang and Möller, 1995; Kono et al., 1998; Takeuchi present in nature as metallic mercury, mono- and bivalent
et al., 1995). It has also been conclusively demonstrated that inorganic compounds, and organic alkyl, aryl, and alloxy-
lupus-prone strains are particularly sensitive to the induc- alkyl compounds. Three main forms of mercury are found in
tion of SLE-like disease following exposure to mercury the environment: elemental mercury, or quicksilver (Hg0),
(Hultman et al., 1992; Takeuchi et al., 1995; Hanley et al., inorganic mercury (Hg⫹ and Hg2⫹), and organic methyl-,
2002). ethyl- and phenylmercury. They are widely distributed in
The discovery of potential SLE inducibility on mercury the environment as a result of human activities. Each form
exposure in humans offers the opportunity for comparison has a different solubility, reactivity, and toxicity (Goldman
with data from murine models of SLE. This means that the and Shannon, 2001; Clarkson, 2002). Between 2700 and
identification of potential SLE susceptibility loci in humans 6000 tons of elemental mercury is released per year via
offers the chance to compare data from murine models of natural means to the biosphere through degassing from the
SLE induced by xenobiotics such as mercury. Most SLE earth’s crust and oceans. Industrial wastes and the combus-
susceptibility loci have been mapped in New Zealand hy- tion of fossil fuels add up to an additional 2000 –3000 tons
brid models; at least 12 of which are located outside the of mercury to the environment (ATSDR, 1999).
interval containing the murine major histocompatibility Industrial discharges of mercury to waterways as ele-
complex, H-2. Three regions commonly noted by linkage ment or inorganic salts have been common practice. It was
studies in New Zealand models are found on murine chro- universally thought that these discharges were innocuous
ENVIRONMENTAL EXPOSURE TO MERCURY 153

because mercury appeared to sink and remain bound to to be lethal to the consumers (CHA, 2001). Organomercuric
sediments. Later it was discovered that certain microorgan- compounds are vastly more toxic than metallic or inorganic
isms in these deposits are capable of converting metallic mercury because they are lipophilic and can penetrate the
mercury as well as inorganic salts to organomercuric com- blood– brain barrier (BBB) to cause toxicity to the nervous
pounds by methylation process (ATSDR, 1999). Methyl- system (ATSDR, 1999).
mercury (MeHg), which is a common organic form of
mercury, tends to bioaccumulate in successive order in the
aquatic food chain: from plankton and algae to herbivores
Biogeochemical Cycling of Mercury
and finally to the top fish predators such as sharks and
fish-eating marine mammals. A similar sequence exists in Through the processes of biomethylation and bioaccumula-
freshwater bodies, with such species as pike and bass having tion, MeHg finds its way to most species of fish and fish-
some of the highest levels of MeHg contamination. Recent consuming animals (Clarkson, 1997; CHA, 2001). Many
use of the autometallography (AMG) technique by Woshner anthropogenic sources have been identified (Fitzgerald and
and group (2002) confirmed that marine mammals do con- Clarkson, 1991). Chloralkali plants discharge inorganic
centrate inorganic elements, particularly mercury, in their mercury as waste into rivers, lakes, and ocean bays. Paper
tissues to the point of toxicosis. Although this technique is pulp factories likewise discharge a variety of mercury com-
considered semiquantitative (Danscher et al., 1994), using it pounds used as slimicides. Contamination of aquatic sedi-
with beluga whales showed they had significantly higher ments now occurs worldwide because of extensive gold-
hepatic concentrations of total Hg [mean total Hg ⫽ 23.49 mining operations (Cleary, 1990). Large quantities of liquid
and 4.97 ␮g/g wet weight (ww) in liver and kidney, respec- mercury are used to extract the sedimentary gold found in
tively; n ⫽ 24] than did bowhead whales, which had riverbeds. Pure gold is recovered when the mercury is
consistently low Hg levels [mean total Hg ⫽ 0.06 ␮g/g ww; evaporated from the amalgam by heating. It has been esti-
n ⫽ 55]. The disparity in patterns of tissue Hg accumula- mated that more than 130 tons of mercury have been re-
tions between the two whale species was attributed to dif- leased annually into the Amazon basin alone (Cleary, 1990).
ferences in the trophic levels they occupy (Woshner et al., The global cycling of mercury begins with the evapora-
2001). Liver sections of beluga whales exhibited AMG Hg tion of mercury from land and sea surfaces. Volcanoes can
granules with marked zonal distribution, clearly reflecting be an important natural source (Cleary, 1990; Richardson,
initial accumulation around portal triads with progressive 1992). The burning of fossil fuel, especially coal and mu-
dissemination around all 3 lobular zones as hepatic Hg nicipal waste incineration, is a major anthropogenic source
concentrations increased with age. This contrasted with to the atmosphere. Mercury vapor, a chemically stable
similar studies in sledgedogs, which demonstrated greater monoatomic gas, is estimated to have a residence time of 1
Hg-catalyzed AMG granule aggregation in the centrilobular year in the general atmosphere (Clarkson, 2002). It is there-
regions of the liver (Hansen and Danscher, 1995). This fore globally distributed even from point sources. Through
finding suggests the existence of interspecific variation in processes yet to be fully clarified, the vapor is oxidized in
mechanisms of mercury deposition. The outcomes from the upper atmosphere to water-soluble ionic mercury, which
most studies in this field indicating the cellular and intra- is returned to the earth’s surface in rainwater. This global
cellular pattern of AMG granule deposition and the absence cycling of mercury results in the distribution of mercury to
of granules in hepatocytic nuclei suggest that the majority of the most remote regions of the planet.
Hg present was originally in organic form, a finding very Lichens have been utilized as biomonitors of mercury
consistent with other food web sources (Rouleau et al., pollution from gaseous emissions (Richardson, 1992) as
2000). It was also found that macrophages contain aggre- well as from wet and dry depositions (Bargagli et al., 1987).
gates of AMG granules, attesting to the importance of these Lichens take up appreciable amounts of mercury within 4
cells in Hg toxicodisposition (Woshner et al., 2002). It has weeks and accumulate levels close to equilibrium within
been calculated that in belugas a total Hg level of up to 8 –16 weeks (Bargagli et al., 1987). The concentration of
⬃5 ␮g/g ww can be detected in the mesenteric lymph mercury in lichens has been used to make an overall esti-
nodes, yet among ringed seals, nodal total Hg was found to mation of mercury deposition and vapor absorption at point
be minimal (Woshner et al., 2002). The results of these sources over an extended period. Seasonal influences such
studies have led to the belief that the reticuloendothelial as temperature variation and wind direction affect levels of
system might represent the main site for degradation of mercury in lichens. High levels of this pollutant are found in
MeHg to inorganic Hg (Suda and Takahashi, 1990). lichens gathered close to the source, with a near-exponential
The potential for bioaccumulation in aquatic food chains decline in levels within a short distance followed by a
was demonstrated in two outbreaks of human poisoning in slower decline to background levels (Barghigiani et al.,
Japan (Clarkson, 1997). Methylmercury released into a 1990). Barghigiani and his group found that mercury con-
large ocean bay bioaccumulated to such an extent that the centrations in lichens are higher than in soil when ambient
local fish population contained levels of MeHg that proved mercury concentrations are low, but lower in Hg-mineral-
154 TCHOUNWOU ET AL.

ized (cinnabar) areas; mercury concentrations were found to diate release of the EtHg to tissues. Such presumptions
vary also between lichen species (Barghigiani et al., 1990). prompted the idea that adjuvant-containing vaccines were a
This indicates that mercury is stable in soil and that the threat to public health. It was calculated that exposure from
atmosphere is a rich source of mercury absorbed by lichens. a single vaccination dose potentially could exceed federal
Concentrations of mercury at any given location are higher guidelines for the mercury reference dose. Routine immu-
in epiphytic lichens than in mosses and many other life nizations could possibly yield a cumulative dose of up to 75
forms (Loppi and Bonini, 2000). Other investigators, in- ␮g of mercury by 3 months of age and 187.5 ␮g by 6
cluding Steinnes and Krog (1977), pointed out that chlor- months of age. Therefore, the American Academy of Fam-
alkali plants constitute major sources of Hg emissions to the ily Physicians, the Advisory Committee on Immunization
air. Sampling from lakes and rivers around the United States Practices, and the U.S. Public Health Service issued a joint
in 1970 found that about 19% of the waterways are con- recommendation that thimerosal be removed from vaccines
taminated with mercury, with a low median value of less (CDC/AAFP/AAP/ACIP, 2000), and its use as a preserva-
than 0.5 parts per billion (Nriagu and Pacyna, 1988). tive in vaccines for childhood immunization schedules has
been banned.
In the United States phenylmercury compounds have
been used as pesticides to prevent mildew growth on walls
Production and Uses of Mercury
and as preservatives in latex paints to prevent discoloration
Methyl- and ethylmercury compounds were first synthe- from the growth of microorganisms. Phenylmercury and
sized in London in the 1860s (Nriagu and Pacyna, 1988). In EtHg continue to be used as bacteriostatic agents for various
the late 1880s diethylmercury was first used in the treatment topical pharmacologic preparations (Zhang, 1984). Dimeth-
of syphilis, a practice that was later stopped because of the ylmercury is used only in research laboratories. It is highly
toxic properties of this agent. However, early in the 20th toxic and can cause death at low levels of exposure (Nier-
century the discovery of the fungicidal properties of the enberg et al., 1998).
short-chain alkylmercury compounds led to commercial ap-
plications in agriculture. These compounds were especially
effective in treating a plant root disease in wheat caused by HEALTH EFFECTS ASSOCIATED WITH
Telletia triticia. By 1914 many different organic mercury MERCURY EXPOSURE
compounds were in use primarily to prevent seed-borne
diseases of cereals and seed grains (WHO, 1990; Clarkson, Elemental Mercury
1997). The ethylmercury fungicides were used effectively
Physical and Chemical Properties
and safely for several decades. However, a number of
outbreaks of poisoning occurred in some developing coun- Elemental mercury (Hg0) has the following characteristics:
tries; two outbreaks happened in rural Iraq in 1956 and 1960 valence of 1 or 2; atomic number of 80; atomic weight of
from the misuse of ethylmercury toluene sulfonilamide (Je- 200.59; melting point of ⫺38.87°C; boiling point of
lili and Abbasi, 1981). 356.58°C; and density of 13.546 g/mL at 20°C (ATSDR,
Thimerosal, a form of ethylmercury (EtHg), commonly 1999). It is liquid at room temperature and is not well
known under various household names as methiolate, mer- absorbed in the gastrointestinal tract. It has a silvery-white
bromin, or mercurochrome, was routinely used as an anti- shiny appearance and at 20°C has a high vapor pressure,
septic (Hammond, 1971; Tan and Perkin, 2000) and for enough to equilibrate in air to reach concentrations 130
topical applications (Pier, 1975). It was also used as an times the maximal industrial allowance of 0.1 mg/m3 (Hu,
effective vaccine preservative and as biological agents for 2000). Only 6 ␮g of mercury in the air is required to reach
medical therapy as well (Zhang, 1984). The EtHg in thimer- ASTDR’s minimal risk level of 0.2 ␮g/m3 in a 10-ft2 (ca. 30
osal undergoes a fast dissociation in the presence of endog- m3) room. Depending on individual susceptibility and ex-
enous thiol groups within cells and tissues. Research has posure time in such a room, health problems become ap-
shown that the types of anions attached to the alkylmercury parent at a mercury volume of between 40 and 400 nL of
radical make little difference to its ultimate disposition in vaporized liquid. Hence, it is a potent, toxic biohazard at
the body (ATSDR, 1999). These include anions such as room temperatures. Inhaled vapor easily crosses the pulmo-
hydroxyl-, cyanide-, and even thiol-containing propane di- nary alveolar membranes to enter the circulatory system,
olmercaptides. Such findings suggest that mercury radical where it invades primarily red blood cells, the central ner-
rapidly dissociates from anions in the parent compounds to vous system, and the kidneys (Clarkson, 1997). Its partial
attach to ligands in tissues, which may partly indicate the conversion to HgCl2 allows it to be retained in the kidneys
role of the methyl group in the high lethality associated with and the CNS for years. Outside occupational settings expo-
MeHg poisoning (ATSDR, 1999). The discovery of these sure to metallic mercury results from broken fever ther-
properties of mercury compounds led to the assumption that mometers, but the amount of mercury liberated is usually
the administration of thimerosal also resulted in the imme- small (0.5–2 g mercury, ⬍ 0.15 mL; Tominack et al., 2002).
ENVIRONMENTAL EXPOSURE TO MERCURY 155

Large quantities of elemental mercury can be spilled in inorganic mercury in humans (Dales et al., 1971; Tan and
school science laboratories, and the attractive nature of Perkin, 2000).
mercury invites pilfering. The greatest biohazard, however,
lies in occupational exposure from the heating procedures
used for its extraction from mercury-rich ores such as cin- Inorganic Mercury Compounds
nabar (ATSDR, 1999).
Physical and Chemical Properties
Some of the most important mercury salts are mercurous
Clinico-Pathological Effects
chloride (Hg2Cl2, or calomel), occasionally used in medi-
Inhalation of volatilized vapors of metallic mercury leads to cines; mercuric fulminate [Hg(OCN2)], a detonator in ex-
chemical pneumonitis. Exposure to high levels produces plosives; and mercuric sulfide (HgS, or vermillion), a high-
acute necrotizing bronchitis and pneumonitis with symp- grade paint pigment (Klassen, 1990). Some inorganic
toms of cough, dyspnea, and chest tightness. Diffuse infil- compounds have been used for generations for antibacterial,
trates seen in early chest radiographs progress to pulmonary antiseptic, cathartic, and diuretic purposes. They also have
edema, respiratory distress, and desquamation of bronchio- been exploited in a number of consumer products ranging
lar epithelia that can end in death from respiratory failure. from teething powders to skin-lightening creams. In the
The most common signs of toxic presentation in children United States such uses have been prohibited, but these
are diffuse pruritic rash, believed to be contact dermatitis. products are still available on the world market.
Originally known as acrodyna, this rash is an idiosyncratic Both metallic- and organic mercury– containing com-
hypersensitivity reaction to mercury at onset of the exposure pounds are oxidized to inorganic mercury in the liver,
(Mathieson et al., 1980; Langford, 1999). It develops into a preferentially utilizing hydroxyl radical production. The
painful, poorly defined erythematous papulovesicular erup- intestinal flora can also convert organic mercury to its
tion, with edema and induration of the palms, soles, and inorganic form (Suda and Hirayama, 1992). However, or-
face. This progresses over a period of weeks into desqua- ganic mercury is not transformed appreciably in the body.
matous ulceration, often accompanied by diaphoresis and Rats and mice treated parenterally with HgCl2 have been
increased heart rate and blood pressure. Enhanced hypoto- found to exhale metallic mercury vapor (Hammond, 1971;
nia, itching, burning and severe pain of the extremities, Rowland et al., 1980). Although only about 10% of ingested
alopecia, ptyalism or profuse secretions, insomnia, apathy, mercury salt is absorbed, it tends to be extremely caustic to
and irritability have been observed in children. Painful back the GI tract. A small amount of dermal absorption occurs as
spasms and urination are also common complaints (Clark- well. In adults the half-life of ingested mercuric salt is about
son, 1997). Poor sleeping, sore throat, night sweats and 40 days. Excretion is mostly fecal. Urinary excretion is
sweating when not hot are noted symptoms. Pustular follic- negligible, on the order of 10% or less of total elimination
ulitis develops later, followed by a painful groin; severe from the body (Clarkson, 2002), but with chronic exposure
constipation and impotence in adults have also been ob- urinary excretion is somewhat greater (Dales et al., 1971;
served. By contrast, less than 0.1% of elemental mercury is Tan and Perkin, 2000).
absorbed from the gastrointestinal tract when ingested It is known, however, that elemental mercury crosses the
(Mathieson et al., 1980). placenta and concentrates in the fetus of a pregnant woman
In contact with tissue and in inhalation, elemental mer- (Pier, 1975; Koos and Longo, 1976; Aschner et al., 1997);
cury is oxidized to its mercuric ion (Hg2⫹) by the catalase its conversion to inorganic forms or to making combinations
enzyme within the blood (Pier, 1975). Mercuric ions do not with sulfur compounds can occur in vivo (Koos and Longo,
cross the blood– brain barrier (BBB) very well. This means 1976; Aschner et al., 1997). Studies among pregnant Swed-
they also diffuse less out of the brain. Long-term exposure ish women indicated a high risk to the fetus. While under-
to mercury vapor primarily leads to CNS defects (Aschner going dental amalgam treatment in the course of antenatal
et al., 1997). Early nonspecific signs including insomnia, management, these women were monitored for exposure to
forgetfulness, loss of appetite, and mild tremor may be inorganic mercury (I-Hg) until delivery. In that same period
misdiagnosed as psychiatric illness. Continued exposure the dietary intake of freshwater fish, a possible source of
leads to progressive tremor and erethism, a syndrome char- bias, was found to be very low and a negligible source of
acterized by an intention tremor, excitability, memory loss, mercury intoxication (Ask et al., 2002). The results indi-
insomnia, timidity, and sometimes delirium, once com- cated that the median concentration of I-Hg in the placenta
monly seen in workers exposed to mercury in the felt-hat at the time of delivery was 4 times higher (up to 7 ␮g/kg
industry, giving rise to the expression “mad as a hatter” b.wt) than that in the maternal blood circulation, and it
(Bittner Jr. et al., 1998). Red palms, emotional lability, increased considerably with an increasing number of dental
salivation, excessive sweating, and hemoconcentration are fillings. The implication is that I-Hg that bound to the
accompanying peripheral and autonomic signs. The urine placenta probably originated from Hg0 released from amal-
and feces are the main excretory pathways of metallic and gam fillings and oxidized to Hg2⫹ by catalase in the blood.
156 TCHOUNWOU ET AL.

The Hg2⫹ ion may bind to metallothionein that is rich in son et al., 1980; Elferink, 1999). Methylmercury is the
cysteine in the placenta (Aschner et al., 1997). In vitro best-known organic mercurial because it is the predominant
studies suggest that Hg2⫹ has an effect on the transfer of form of organic mercury found in the environment and has
amino acids (Kerper et al., 1992), on placenta oxygen con- been known to be toxic for centuries (Goyer, 1996). Con-
sumption (Aschner et al., 1997) and enzyme activity, and on sumption of contaminated fish is the primary route of hu-
hormonal secretion (Boadi et al., 1992; Kerper et al., 1992). man exposure to organic mercury. Methylmercury appears
Inorganic mercury also accumulates in kidneys, and occu- in human milk. In the blood MeHg has a mean half-life of
pational exposure can cause renal damage. Amalgam fill- 40 –50 days (range: 20 –70 days) for adults from atmo-
ings and diet are the main sources of exposure in the general spheric inhalation or ingestion. Ninety percent of MeHg is
population. excreted in feces through bile (Zhang, 1984). Phenylmer-
cury is rapidly metabolized. Its effects are similar to those of
Clinicopathological Effects mercury salts (Suzuki et al., 1973; Urbach et al., 1992;
Bittner Jr. et al., 1998; Hu, 2000).
The toxic effects of mercurials are universally dependent on
the degree of solubility and specifically rely on the ability to
combine with functional sulfhydryl (-SH) groups in impor- Clinicopathological Effects
tant enzymes. The principal syndrome of acute mercury salt It has been known for centuries that methylmercury is toxic
poisoning is stomatitis, or digestive upset. Mercury salts are (Mathieson et al., 1980; Sager et al., 1984; WHO, 1990). In
very toxic to the kidneys, causing acute tubular necrosis, addition, it has been reported to be neurotoxic, particularly
immunologic glomerulonephritis, or nephrotic syndrome. in children. It inhibits in vitro microtubule formation and
Hence, excessive exposure leads to renal injury. Chronic protein synthesis in neurons, alters membrane activity, and
exposure primarily involves the CNS, ultimately causing disrupts DNA synthesis (Goyer, 1996). Within cells it im-
permanent damage. Symptomatologies of chronic toxicity pairs mitosis and interferes with neuronal migration (Goyer,
include progressive anemia, gastric disorders, salivation, 1996). Prenatal or postnatal exposure to MeHg adversely
metallic taste in the mouth, inflammation, and tenderness of affects the CNS but seems to be most neurotoxic prenatally
gums and tremors (Urbach et al., 1992; Hu, 2000). Central in the course of brain development (Choi et al., 1978;
neuropathy can also occur from mercury salt exposure. Albers et al., 1988). Exposure to high levels of MeHg
Acrodynia (painful extremities) was also reported among causes mental retardation, cerebral palsy, seizures, and ul-
infants exposed to calomel teething powders containing timately death (WHO, 1990; Bittner Jr. et al., 1998). The
mercurous chloride (Mathieson et al., 1980; Suzuki et al., toxicity of organic mercury compounds depends on the
1973). Darkening and loss of teeth may be the final toxicity specific compound, route of exposure, dose, and age of a
associated with some mercury compounds. Common symp- person at the time of exposure. The overt clinical effects
toms, however, are renal complications, with extensive al- resulting from toxic exposure to mercury have been de-
terations in behavior (Suzuki et al., 1973; Bittner Jr. et al., scribed. Fatal ingestion usually is either inadvertent or with
1998). suicidal intent. Gastrointestinal ulceration, perforation, and
hemorrhage rapidly occur, followed by circulatory collapse.
Breakdown of intestinal mucosal barriers leads to extensive
Organic Mercury Compounds mercury absorption and distribution to kidneys, causing
severe kidney damage (Eley, 1997; Goldman and Shannon,
Physical and Chemical Properties
2001; Clarkson, 2002; Hanley et al., 2002; Hultman et al.,
Naturally occurring mercury derives from the divalent form, 1998; Pollard et al., 2002).
Hg2⫹ which is more stable in association with sulfur in the The nervous system is more sensitive to mercury toxicity
mineral ore cinnabar and in fossil fuels such as coal and than any other system in the body. Mercury has also been
petroleum. Once discharged into the atmosphere through associated with arrhythmias and cardiomyopathy. Hair anal-
mining and industrial activities, mercury undergoes a vari- yses of patients with mercury-induced cardiomyopathy
ety of transformations (Tan and Perkin, 2000; Goldman and measured mercury levels 20,000 times higher than in con-
Shannon, 2001). Bacteria in lakes, streams, and ocean sed- trols (Harada et al., 1998; Frustaci et al., 1999). Other
iment can convert elemental mercury to its organic com- neurological problems such as tremors, insomnia, polyneu-
pounds, which may accumulate in fish and in the food chain ropathy, paresthesia, emotional lability, irritability, person-
(Clarkson et al., 1988; Langworth et al., 1991; CHA, 2001). ality changes, headache, weakness, blurred vision, dysar-
The methyl, ethyl, and phenyl series are the main organic thria or speech impairment, slowed mental response, and
forms that have been marketed as industrial compounds, unsteady gait have been observed (Chang et al., 1995;
primarily as biocides and as pesticides. They are also found Marsh, 1995; Myers et al., 1995; Urbach et al., 1992; Chan,
in two once-common household antiseptics: mercuro- 1998). Many cases have been reported of infants exposed to
chrome (merbromin) and merthiolate (thimerosal; Mathie- phenylmercury used as fungicidal diaper rinse and of chil-
ENVIRONMENTAL EXPOSURE TO MERCURY 157

dren exposed to mercury in interior latex paint (Agocs et al., murine macrophages and dendritic cells; this supports the
1990; Langford and Ferner, 1999). A maculopapular rash, idea that HgCl2-induced cell death (HgICD) contributes
swollen nodes and painful extremities, peripheral neuropa- immunogenic complexes capable of breaking immunologi-
thy, hypertension, and renal tubular dysfunction developed cal self-tolerance (Biancone et al., 1996).
in affected children (ATSDR, 1999). However, individual
susceptibility to organomercury is poorly understood.
Mercury-Induced Antibody Synthesis
Thimerosal, an ethyl derivative of mercury, has been in
use for many decades, but very little information exists in Hirsch and colleagues (1982) isolated a range of antibodies
the literature regarding its toxicological effects. Most esti- after injecting Brown-Norway (BN) rats with 0.1 mg of
mates of health risks from thimerosal found in vaccines HgCl2/100 g of body weight 3 times weekly for variable
stem from extrapolations from findings with methylmercury periods ranging from 11 to 25 days. The antibodies detected
(CDC/AAFP/AAP/ACIP, 2000; Bell et al., 2001). Dimeth- included antiglomerular monoclonal antibodies (mAbs) of
ylmercury requires specialized storage and handling be- IgM isotype mAbs of IgE, mAbs against mitochondrial
cause of toxicity and penetrating characteristics. Latex antigens, antinuclear antibodies of IgM isotype, IgE mAbs
gloves do not afford an effective barrier, and it is absorbed of no detectable specificities, and others that reacted with
through the skin. Exposure to only a 400-mg dose can lead single-stranded DNAs (ss-DNA; Hirsch et al., 1982). Other
to degeneration in the CNS, particularly in the cerebellum, investigators have confirmed that mercury can induce a
to end in death (Nierenberg et al., 1998). Despite the noted large spectrum of autoantibodies through polyclonal B-cell
toxic effects of MeHg, there is no consensus among health activation (Frustaci et al., 1999). Features associated with
professionals as to what constitutes a safe level of exposure HgIA are not dissimilar from the manifestations of SLE;
(Bell et al., 2001). these include lymphocyte proliferation (Hirsch et al., 1982),
high association with specific class II MHC gene expression
(Dubey et al., 1991), hypergammaglobulinemia (Bariety et
al., 1971), polyclonal Abs to self Ags, production of anti-
IMMUNOTOXIC EFFECTS OF MERCURY
nuclear Abs, AnoA Abs (Bariety et al., 1971; Hirsch et al.,
1982; Dubey et al., 1991), ICGN (Bariety et al., 1971), and
Mercury-Induced Immune Responses
necrotizing vasculitis (Mathieson et al., 1992). Autoanti-
Many metals, including mercury, have potent cytotoxic bodies similar to those that lead to glomerulonephritis in
effects on specific types of cells, including those of the humans have been found in drug and substance abusers
immune system. Exploitation of the cytotoxic property of (Clarkson, 1972; Pollard et al., 1999). Most findings on the
metals in medicine has been very well utilized in the past, role of mercury in such immune reactions have been in the
especially in the management of rheumatoid arthritis (gold murine species (Biancone et al., 1996; Pollard et al., 1997;
therapeutic formulations) and in the early treatment of syph- Kono et al., 1998; Pollard et al., 1999; Hanley et al., 2002;
ilis. Mercury is not only capable of inducing immunosup- Pollard et al., 2002), however, and clinical manifestations of
pression but is also able to produce immunostimulatory HgIA are also similar to spontaneous SLE in humans
signals in many species, humans and rodents included (Pol- (Hirsch et al., 1982).
lard et al., 1999). Exposure to mercury causes cell death by For induction of the autoimmune state, it has been de-
a nonapoptotic process primarily based on cellular morphol- termined that T cells, CD40/CD40 ligands, and B7/CD28
ogy (Kass et al., 1990). The prelethal phase imitates oncosis costimulatory molecule interactions are required (Biancone
and occurs within the first 2 h of exposure (Pollard et al., et al., 1996; Kono et al., 1998). HgIA susceptibility is also
2002). Proteolytic degradation of fibrillarin is characteristic strongly influenced by the MHC, with specific class II
of the prelethal phase of HgIA. Fibrillarin is a 34 kDa haplotypes determining autoantibody specificity and disease
protein component of small nucleolar ribonucleoprotein severity (Fillastre et al., 1988; Dubey et al., 1991; Hultman
particles (snRNPs), and its degradation takes place in mac- et al., 1993). Some of the susceptibility genes map to the
rophages. The most likely source of novel fibrillarin cleav- MHC region (Hultman and Enestrom, 1987). RT-1n rats are
age products is phagocytes that accumulate necrotic debris susceptible, whereas rats with RT-11 haplotypes are resis-
after toxic contact with mercury. Mercury concentrates in tant (Sapin et al., 1984; Eneström and Hultman, 1995).
such an environment and causes cell death. Cleavage by cell A.SW mice and others that carry the H-2s haplotype show
proteases perpetuates the rise of new antigens. Further high susceptibility to Hg-induced autoantibodies, whereas
phagocytosis leads to the potential processing and presen- C57BL/6 strains (H-2b) are less susceptible. DBA/2 mice
tation of antigens to T cells to elicit autoimmunity. T-cell strains bearing H-2d haplotypes are not responsive, whereas
proliferation and cytokine release, including IL-1 secretion H-2k-bearing mice show intermediate susceptibility (Dubey
by peritoneal macrophages, following HgCl2 exposure have et al., 1991; Hultman et al., 1993; Jiang and Möller, 1995;
been observed (Biancone et al., 1996). It has also been noted Kono et al., 1998). The AnoA Abs response directed against
that necrotic rather than apoptotic cell debris is activated in fibrillarin is one of the most representative manifestations of
158 TCHOUNWOU ET AL.

HgIA linked to H-2s (Hanley et al., 2002; Hultman et al., jections of HgCl2 in murine species can cause differential
1992; Hultman et al., 1993; Pollard et al., 2002) and, more appearance of TH1/TH2 cell populations, with TH1 high in
specifically, to the class II I-As molecule by analysis of H-2 responders and TH2 increases in nonresponders (Hultman et
congenic mice (Pollard et al., 2002). al., 1992). These TH1/TH2 cells are CD4⫹ T lymphocytes
Other genes are believed to participate in the process, (helpers) that arise in response to antigenic stimulation
which is highly supported by only certain strains of rats and (Sapin et al., 1981; Pietsch et al., 1989). TH1 cells produce
mice being subject to the induction of autoimmune reactions IFN-␥, IL-2, and TNF-␤. They have receptors for IL-12R␤
with mercury (Hultman and Enestrom, 1987). Although on their membrane surfaces; and they promote cellular
several studies have clearly shown genetic susceptibility to immune responses (Kono et al., 1998; Szabo et al., 1997).
autoimmune induction to be partly controlled by MHC- Helper TH2 cells produce IL-4, IL-5, IL-6, IL-10, and IL-13,
linked genes (Sapin et al., 1984; Dubey et al., 1991; Hult- express IL-4-related GATA-3 transcription factor, IFN-␥R
man et al., 1992; Hultman et al., 1993; Jiang and Möller, ␤ (Hultman et al., 1993; Zheng and Flavell, 1997), and the
1995; Biancone et al., 1996), the detailed mechanism of the CCR3 chemokine receptor (Sallustro et al., 1997), and pro-
involvement of another, non-MHC genetic control of re- mote humoral immune responses. Therefore, the dominance
sponsiveness is still speculative. A strong genetic control of one or the other of the TH pathways in an immune
exists for the development of autoimmune manifestations reaction results in either a predominantly cellular (TH1-
both in rats and in mice injected with HgCl2 (Sapin et al., mediated) or a mainly Ab (TH2-mediated) response. Both in
1984; Hultman et al., 1992; Eneström and Hultman, 1995). vitro (Mosmann et al., 1986; Pelletier et al., 1986) and in
However, DBA/2 mice of the H-2d haplotype that do not vivo (Mosmann et al., 1986; Bottomly and Constant, 1997;
respond at all to HgCl2 manipulations because of failure to Hultman et al., 1998) shifts to either of these responses have
generate autoimmune reactions (Hultman et al., 1992; Jiang been reported, although most immune responses involve the
and Möller, 1995; Takeuchi et al., 1995; Kono et al., 1998) participation of both TH1 and TH2 responses, depending on
can still be induced in spleen cultures to yield a low prolif- the immunogen used.
erative response with HgCl2 injections (Hultman et al., Mercury-induced autoimmunity is one of the autoim-
1992). Although BALB/c (H-2d) mice fail to produce anti- mune models in which TH1/TH2 imbalance has long been
nuclear Abs under HgCl2 treatment, they do develop auto- thought to play a critical role (Hirsch et al., 1982; Sapin et
immune reactions measurable by significantly increased al., 1984; Mirtcheva et al., 1989; Dubey et al., 1991; Jiang
levels of Igs in serum and high renal immune complex and Möller, 1995; Biancone et al., 1996). Although the
deposits, including granular mesangial and systemic vessel- mechanism by which mercury modifies the immune system
wall IgG and deposits of C3, the third and central comple- is obscure, it is known that cationic mercury has a high
ment protein (Eneström and Hultman, 1995). This means affinity for sulfhydryl groups as its principal site for binding
that BALB/c mice are actually high responders to mercury and also has a substantial affinity for amines and phospho-
(Hultman et al., 1992). The contradiction is that both DBA/2 ryl, carboxyl, and hydroxyl groups (ATSDR, 1999). Mer-
and BALB/c mice share the same H-2d haplotype. There- cury therefore can form ligands with proteins and can form
fore, their respective opposite susceptibilities to autoim- complexes capable of activating the immune system. Some
mune disorders after HgCl2 injections and different respon- of the modified proteins may have epitopes closely resem-
siveness under in vitro stimulation indicate that genetic bling self-immunogens, easily leading to autoimmune dis-
susceptibility is influenced not only by the MHC loci but orders in predisposed individuals (Takeuchi et al., 1995;
also by the involvement of other genes present outside the Pollard et al., 1997; Pollard et al., 2002). The activation of
MHC complex. The non-MHC background genes obviously CD4⫹ and CD8⫹ T cells requires a prior induction of
differ a lot between the two strains; and the effects of antigen-presenting cells (APC; Jiang and Möller, 1995).
environment need to be elucidated (Hultman et al., 1992). Mercury binds to molecules on accessory APC cells and
Jiang and Möller (1995) determined that the significant transform molecules on these cells to superantigens capable
difference in the in vivo induction of autoimmunity between of activating T cells with a particular set of V␤ Ag-binding
the responder BALB/c strain and the nonresponder DBA/2 receptors (Jiang and Möller, 1995). Mercury-induced auto-
strain could be attributed to the ability of their T-cell sub- immunity therefore utilizes a different mechanism from that
populations to undergo transformations with HgCl2 expo- induced by polyclonal cell activators (PCAs), such as
sure. Only CD8⫹ T cells undergo transformation in DBA/2 pokeweed mitogen (PWM), where T-helper cells are not a
spleen cells on stimulation with HgCl2, whereas both CD4⫹ prerequisite. The presence or absence of IFN-␥ on re-
and CD8⫹ T cells enlarge in BALB/c spleen cells (Hultman sponder or nonresponder TH1 and TH2 cell types, respec-
et al., 1992; Jiang and Möller, 1995). Transformed TH cells tively, is thought to be a necessity for whether the response
play critical roles in the development of autoimmunity occurs or does not occur, respectively (Kono et al., 1998). It
induced by mercury; and these helper TH cells may further is contended that the balance between TH1 and TH2-type
assist in inducing B cells to produce autoantibodies and responses does not contribute directly to autoimmune sus-
finally immune complex-mediated glomerulonephritis. In- ceptibility. IFN-␥ is required for the activation of the im-
ENVIRONMENTAL EXPOSURE TO MERCURY 159

mune system to respond to poor antigenic determinants, pared with naive mice, these mice did not show any decline
including both self and nonself Ags involving humoral and in the number of spleen plaque-forming antibody-secreting
cellular autoresponses. Differences in the dose levels of cells and/or diminution of serum titers of specific IgM,
IFN-␥ appear to contribute directly to disease susceptibility. IgG1, and IgG2 anti-HRBC antibodies in response to
Immunization with high doses of Ag and a strong adjuvant HRBC (Hanley et al., 1997). Similarly, in mercury-resistant
tend to bypass the IFN-␥ requirement (Ferber et al., 1996; DBA/2 (H-2d) mice, chronic treatment with mercury did not
Jones et al., 1997). Similarly, a strong genetic predisposition suppress total specific antibody responses against HRBC.
may decrease the threshold for susceptibility enough to Immunosuppression appeared to be under MHC gene con-
outweigh the IFN-␥ requirement (Abbas et al., 1996). Sus- trol because there was no general immunosuppression;
ceptibility to autoimmune disease therefore is a multistep rather, it was found to be an intrinsic property of B cells
process with many stages or checkpoints, considering the (Hanley et al., 1997, 2002). This was reinforced by recent
clinical findings in SLE patients (Andre et al., 1996; Hult- findings that F1 generation of hybrid mice carrying the s and
gren et al., 1996; Manoury-Schwartz et al., 1997; Vermeire b haplotypes are resistant to HgIA because they fail to
et al., 1997). The IFN-␥ requirement for autoantibody pro- synthesize antifibrillarins, which are antinucleolar autoanti-
duction, as an example, defines a checkpoint at the initiation bodies (AnoAbs) induced with mercury injections (Hanley
of the autoimmune process with the potential to distinguish et al., 2002). The F1 generations were the result of crosses
self from nonself. Understanding the complex lupus genes between sensitive H-2s and resistant H-2b haplotypes. The
by probing into the biology and mechanisms of lupus patho- results contradicted theoretical expectations because the I-A
genesis will certainly provide important opportunities for molecule is codominantly expressed in F1 mice; and in other
enhancing diagnostic procedures, therapeutic treatments, autoimmune models heterozygosity of class II was found to
and the possible means of preventing this genetically com- enhance autoimmunity or moderately affect Ab titers (Nyg-
plicated autoimmune disorder. Murine studies that have ard et al., 1993; Hultman et al., 1995a,b). The F1 hybrids of
induced autoimmunity with mercury therefore have a pro- the H-2s/b strains were found to be resistant to AnoAb
pensity to contribute to the understanding of the self/nonself production when injected with mercury. From results of
recognition system within the range of immune responses. adoptive bone marrow transfer studies this resistance to
mercury appeared to be an intrinsic characteristic of H-2s/b
(heterozygous) B cells in relation to AnoAb production
Mercury-Induced Immunosuppression
(Hanley et al., 2002).
A peculiar feature of HgIA is that polyclonal B-cell activa- In the DBA/2 strain of mice that are resistant to HgIA
tion spontaneously disappears despite continuous injection despite expressing a susceptible H-2 (H-2d) haplotype, for
of mercury (Roether et al., 2002). The mechanism or mech- example, the expression of the Hmr1 locus genes has been
anisms are not well clarified for autoregulation of HgIA in shown to be very complex. The Hmr1 locus, a single major
susceptible rats and mice. HgIA normally has the charac- quantitative trait locus on chromosome 1, contains several
teristics mentioned previously, that is, T-cell-dependent lupus susceptibility genes (Kono et al., 2001). The com-
polyclonal B-cell activation that culminates in increased plexity is thought to be an intrinsic property of the B cells’
serum levels of immunoglobulin (IgG1 and IgE), produc- resistance to HgIA (Hanley et al., 2002). This information
tion of antibodies of different specificities, and development supports that non-MHC genes are involved in HgIA because
of renal IgG deposits. In testing for and analyzing the source the genes for MHC expression in mice are on chromosome
of the spontaneous downregulation of mercury-induced im- 17. It has also been shown that epistatic interaction of the
mune/autoimmune responses, it has been found that the Sle1, also found on chromosome 1 of the New Zealand
inhibition of HgIA is not a result of general immunosup- model of SLE, and FASlpr, the CD95 genes, has an additive
pression because the experimental animals were still capa- production of lymphosplenomegaly with increased numbers
ble of responding to different or unrelated antigens (Roether of activated CD4 T cells, CD4⫺CD8⫺ double negative
et al., 2002). (DN) T cells, and B1a cells and high levels of IgG and IgM
Mercury-susceptible SJL (H-2s) and -resistant DBA/2 antinuclear (including anti-ssDNA, anti-dsDNA, and anti-
(H-2d) mice were injected with mercury for 4, 10, 15, and histone/DNA) and antiglomerular autoantibodies (Shi et al.,
17 weeks. Immune and autoimmune responses were moni- 2002). The Sle1 itself is a potent locus that triggers the
tored in these mice. Thereafter, the mice injected with formation of IgG antihistone/DNA antibodies when ex-
mercury for 17 weeks were further immunized with horse pressed on the B6 background as a congenic interval (Shi et
red blood cells (HRBC) in order to monitor the subsequent al., 2002). Above all, histological and clinical evidences of
humoral immunosuppressivity if any, to a foreign antigen. glomerulonephritis induced in NZM2410 strains of mice
Except for IgG1 antinucleolar antibody production and re- resulted in more than 80% being fatalities within 5– 6
nal IgG1 deposition, other characteristics of HgIA were months; this confirms the presence of non-MHC genes with
downregulated in the SJL (H-2s) mice after chronic treat- the added influence of susceptibility and resistance genes to
ment with mercury (Roether et al., 2002). However, com- HgIA (Stiller-Winkler et al., 1988; Pollard et al., 1997). It is
160 TCHOUNWOU ET AL.

highly probable that xenobiotic compounds in general and occurs when there is exposed to many stimuli, including
mercury in particular induce autoimmunity in genetically high levels of mercury and UV light or viral infections.
predisposed individuals. More investigations are needed to Apoptosis results in nuclear fragmentation and the accumu-
define genetic alterations and mechanisms responsible for lation of nuclear autoantigens into cell surface blebs, or
the Hmr1 phenotype in order to deduce the relationship apoptotic bodies (Casiano et al., 1996) and ends up in the
between the environment and genetic susceptibility in- release of intracellular immunogens to the surrounding tis-
volved in autoimmune diseases. sue, which leads to self-Ag presentation and autoimmunity.
A marked increase in IL-4 production is also a known Apoptotic bodies contain several self-epitopes, as well as
feature of HgCl2-treated mice (Ochel et al., 1991; Van Vliet many components comprising autoantigenic macromolecu-
et al., 1993; Gillespie et al., 1995). T-cell help is required in lar complexes (Pollard et al., 1997; Pollard et al., 2002), and
this model because nude mice with an H-2s background fail these molecules are targets for multiple autoantibody re-
to develop autoantibodies (Hultman et al., 1992, 1995a). sponses. But apoptosis is believed to be a noninflammatory
High levels of IL-4 messenger RNA (mRNA) have been process (Pollard et al., 2002), and the processing and pre-
detected in the splenic CD4⫹ T cells of HgCl2-treated H-2s sentation of self-Ags as a result of apoptotic cell death
mice, whereas those of H-2d mice showed only a weak probably contribute more to deletion and tolerance than to
increase (Van Vliet et al., 1993). Treatment of H-2s mice stimulation of autoreactive cells (Roether et al., 2002).
with anti-IL-4 mAbs did not stop AnoAb production but Autoimmunity therefore is considered more likely to be
only altered the isotypes of the autoantibodies (Ochel et al., associated with defective PCD, such as the acceleration of
1991). Despite possible noncognate help via cytokines, it systemic autoimmunity found in mice with the lpr or gld
has been demonstrated that B cells require specific MHC mutations (Pollard et al., 2002). That does not exclude
signals from T cells to produce AnoAbs (Hanley et al., nonapoptic cell death, and the dose of mercury plays an
2002). Lack of responsiveness in HgIA is therefore a prob- important role as well.
able intrinsic regulatory property of B cells because of CD95, also called Fas, is a transmembrane protein that is
MHC-guided determinant capture arising from MHC mol- a part of the tumor necrosis factor receptor (TNFR) super-
ecules competing for determinants in APC Ag processing family, whose members are defined by having cysteine-rich
(Sercarz et al., 1993). extracellular domains and a homologous cytoplasmic se-
quence termed the “death domain,” capable of interacting to
bring about PCD and/or the mediation of functions that are
distinct from or even that counteract apoptosis (Ashkenazi
Mercury-Induced Immune Responses
and Dixit, 1998). Dysregulation of Fas-mediated apoptosis
and Apoptosis
is a known factor contributing to the rise of autoimmune
Abnormal accumulation of autoreactive lymphocytes be- disorders (Takahashi et al., 1994; Casiano et al., 1996).
cause of defective termination of lymphocyte activation and Some of these proteins are death receptors (DR) associated
growth via apoptosis characterizes numerous systemic au- with the initiation of apoptosis (Ashkenazi and Dixit, 1998).
toimmune diseases (Theofilospoulos and Kono, 1999). The importance of CD95 in immunoregulation is evidenced
Studies of rodents have clearly indicated a link between by the homozygous CD95 defect in the lpr and gld genes in
mercury exposure and autoimmune disease. Human case mice. Homozygous mutations of the Fas or FasL genes,
reports have connected accidental mercury contamination affect Fas-induced PCD and cause respectively, lymphopro-
and autoimmunity (Leonard et al., 1983a; Röger et al., liferation and a generalized autoimmune disease state.
1992; Pollard and Hultman, 1997). The characteristic fea- MRL⫹/⫹ mice have autoimmune proliferative syndrome
tures associated with HgIA in rodent models have already indicated by massive lymphadenopathy and lupuslike im-
been mentioned. Lymphoproliferation, autoreactive CD4⫹ munopathology (Takahashi et al., 1994). The pathogenesis
T-cell generation, T-cell-dependent polyclonal B-cell acti- of the disease is more severe and detected early in MRL
vation, hypergammaglobulinemia, high serum IgE levels, lpr/lpr mice, those with the homozygous CD95 defects
and the synthesis of autoantibodies that proceed on to im- (Watanabe-Fukunaga et al., 1992; Takahashi et al., 1994).
mune complex–mediated tissue injury and glomerulone- Similarly, humans who have CD95 mutations with an lpr-
phritis have all been detected (Bariety et al., 1971; Roman- like genetic constitution also suffer from a variety of lym-
Franco et al., 1978; Hirsch et al., 1982; Leonard et al., phoproliferative autoimmune diseases (Fisher et al., 1995;
1983a; Röger et al., 1992; Theofilospoulos and Kono, 1999; Le Deist et al., 1996; Dianzani et al., 1997; Puck and
Shi et al., 2002). Sneller, 1997; Whitekus et al., 1999). These patients also
The mechanism or mechanisms linking mercury and have defects downstream in the CD95 death pathway. CD95
autoimmunity are poorly understood. Normally cell death and CD95L also are implicated in pathological suppression
occurs after proteolysis of intracellular proteins, including of immune surveillance, that is, elimination of tumor-reac-
nuclear autoantigens. Apoptosis, programmed cell death tive immune cells by certain tumors that constitutively
(PCD), is a morphological description of cell death that express CD95L. Thus, defects in the apoptotic-signaling
ENVIRONMENTAL EXPOSURE TO MERCURY 161

pathway appear to be linked to autoimmune disease suscep- tween nonapoptosis (HgCl2, heat, ethanol) and apoptosis-
tibility. (anti-Fas) induced cell death (Casiola-Rosen et al., 1995;
Mice injected with low concentrations of mercuric chlo- Pollard et al., 1997). From such studies it is becoming
ride (⬍ 40 ␮M and definitely ⱕ 10 ␮M) end up with apparent that an MHC-restricted autoantibody response and
disruption in the CD95-mediated apoptotic cell death path- interaction with HgCl2 are characteristics that differentiate
way (Whitekus et al., 1999). Mercury has been found to fibrillarin as an autoantigen in HgCl2-induced autoimmu-
attenuate CD95-mediated apoptotic cell death; the molecu- nity. The observation that specific cleavage fragments of
lar target for mercury appears to localize downstream of fibrillarin result from HgCl2-induced death and not other
agonist binding to CD95 and upstream of caspase-3 activa- forms of cell death means that novel cleavage fragments
tion. Mercury as Hg2⫹ with its high affinity for free protein probably act as autoimmunogens. In addition, other effects
sulfhydryls (Clarkson, 1972) can associate with critical thi- of mercury on the immune system including specific cyto-
ols in CD95; by so doing it is likely to inhibit agonist kine requirements (Ochel et al., 1991; Van Vliet et al., 1993;
binding and subsequently affect signal transduction events Gillespie et al., 1995) and inhibition of Fas-mediated cell
(Whitekus et al., 1999). But Hg2⫹ is not known as a com- death are possible means of terminating self-tolerance, lead-
petitive inhibitor of CD95 binding, nor does it interfere with ing to the equivalent of the SLE state (Whitekus et al.,
the TNF-/TNF-R interaction (Casiano et al., 1996). The 1999).
functional importance of the cysteine-rich sequences in the It was long ago determined that in humans the predom-
extracellular domains of both CD95 and TNF-R is believed inant characteristic of SLE is formation of antinuclear an-
to depend on receptor trimerization required for the appro- tibodies, particularly against double-stranded DNA
priate propagation of the death signal (Ashkenazi and Dixit, (dsDNA; Berden, 1997). This autoimmune response is T-
1998). Like other TNF family members, CD95L is a ho- cell- and (auto)-antigen dependent. But dsDNA is very
motrimeric molecule, and crystal structure studies of its weakly immunogenic. It has been found, however, that the
complexed forms with TNF1 suggest that each CD95L nucleosome is the principal autoantigen in SLE and that
trimer binds three CD95 molecules (Ashkenazi and Dixit, DNA circulates in SLE patients as nucleosomes. Nucleo-
2002), leading to clustering of the receptor’s death domains, somes in vivo, on the other hand, are generated through
which initiates the death process. apoptosis. There is also increasing evidence that apoptosis is
Pollard and coworkers (1997, 2002) demonstrated that disturbed in human SLE patients (Berden, 1997). Nucleo-
mercury-induced cell death (HgICD) led to a proteolytic somes have a high affinity for heparin sulfate in the glo-
cleavage of fibrillarin that was specific to this xenobiotic; merular basement membrane (GBM). Through complexes
but the generation of a unique (19 kDa) proteolytic cleavage with nucleosomes, antinuclear antibodies (both nucleo-
fragment did not need interaction between mercury and some-specific and anti-dsDNA autoantibodies) acquire a
fibrillarin. Instead, HgICD was associated with a novel high affinity for GBMs. This is an initial key event in the
protease activity not detected in other forms of induced cell development of lupus nephritis. It has been demonstrated
death. It was found that xenobiotic-induced cell death pro- that the pentapeptide Asp/Glu-Trp-Asp/Glu-Tyr-Ser/Gly is
duced novel protein fragments that stimulated self-reactiv- a molecular mimic of dsDNA (DeGiorgio et al., 2001). This
ity, quite different from that elicited by the full-length sequence is also found in the extracellular domain of murine
protein (Pollard et al., 2002). Above all, xenobiotic-induced and human NMDA (N-methyl-D-aspartate) receptor sub-
autoimmunity characterized by autoantibody responses units NR2a and NR2b. The NR2 has been shown to cross-
against native self-Ag did not require preinteraction be- react with both murine and human anti-DNA antibodies to
tween xenobiotic and Ag. The genetically restricted antifi- mediate the apoptotic death of neurons in vivo and in vitro
brillarin autoantibody response of HgIA was not found to be (Berden, 1997). Lupus antibodies cross-react with DNA and
directed against a fibrillarin–Hg complex, although a metal– NMDA receptors, cross the BBB, and probably mediate
protein interaction occurred (Pollard et al., 2002). This nonthrombotic and nonvasculitic abnormalities of the cen-
finding provides evidence buttressing a longstanding belief tral nervous system (Berden, 1997).
that SLE-prone patients could generate autoantibodies spon-
taneously, even without any physical presence of observ-
able inducers of autoantigens (Pollard et al., 1997, 2002). Significance of Mercury-Induced
From further studies with HgCl2 and VP-16, which is an Autoimmunity
apoptosis-inducing etoposide, it was demonstrated that cell
Systemic Autoimmunity
death occurs via both nonapoptotic and apoptotic activities
of proteases that were sensitive to the same inhibitors; but Autoimmune diseases may appear as organ-specific or sys-
the cleavage patterns of fibrillarin were different enough to temic pathology affecting all tissues of the body, particu-
suggest the action of different proteases (Casiano et al., larly the kidneys. Both forms can be associated with nu-
1996; Pollard et al., 1997; 2002). This indicates that the merous immune pathologies (Theofilopoulos, 1995; Aten et
cleavage patterns for a number of autoantigens differ be- al., 1998). Epidemiological data have indicated that SLE
162 TCHOUNWOU ET AL.

affects 15–50 of every 100,000 people. The male-to-female al., 1982; Theofilospoulos and Kono, 1999). The mecha-
ratio is 3:1 (Hahn, 1994). Between 40% and 50% of SLE nisms of susceptibility and resistance, though, are still eva-
patients, compared to 15% in many healthy ethnic control sive.
groups, have a defective C4AQO, a HLA class III allele that
fails to encode the C4A protein, necessary for the function
Pathogenesis of Autoimmunity by Mercury
of the complement system of proteins (Arnett Jr., 1993;
Hahn, 1994). In some populations specific haplotypes, Research has shed more light on the mechanisms whereby
B8.DR3.DQ.2 and C4AQO, predispose people to SLE. The mercury induces antibodies against self-antigens. Subtle
strongest association involves the class II DQB for which differences in the type of immune response generated by
high titers of IgG anti-DNA are associated with lupus ne- murine species seem to depend on the dose, form, and route
phritis, especially in such combinations as DQ␤1*0201, of administration of Hg (Hirsch et al., 1982; Pollard et al.,
0602, and 0302 with DR2 or DR3 (Arnett Jr., 1993). Anti- 1997; Pollard et al., 2002). Mercuric chloride (HgCl2) salt
bodies to Ro/La (SS-A/SS-B) are associated with dermatitis administered subcutaneously at levels of 40 ␮g of HgCl2 in
of subacute cutaneous lupus and with certain DQA and DQB PBS twice a week for 10 weeks in SJL/J mice induced the
genes inherited with DR3 and occasional DR2 (Arnett Jr., formation of autoantibodies to fibrillarin (Pollard et al.,
1993). 2002).
Twin studies of SLE patients have confirmed that al- Analyses of the autoantibody responses to fibrillarin in
though genetic predisposition may be required for the de- murine models of autoimmunity against fibrillarin are sim-
velopment of the disease, environmental exposures are im- ilar to antifibrillarin responses in a subset of human sclero-
portant for its full expression (Deapen et al., 1992; Pollard derma disease (Guery et al., 1990; Deapen et al., 1992;
et al., 2001). In both animal models (Hirsch et al., 1986; Kasturi et al., 1995; Takeuchi et al., 1995) that also show
Pietsch et al., 1989; Guery et al., 1990; Mathieson et al., genetic restriction (Arnett et al., 1996). It is mapped to the
1992; Hultman et al., 1994; Kono et al., 1994; Arnett et al., H-2A region of the MHC complex of H-2s mice (Pollard et
1996; Aten et al., 1998; Pollard et al., 1999) and epidemi- al., 2002; Shi et al., 2002). These antibodies arise via B-cell
ological studies (Deapen et al., 1992; Theofilopoulos, 1995; responses (Pollard et al., 1999). This finding assists in
Pollard et al., 1997) environmental agents such as mercury probing into the mechanism whereby HgCl2 elicits an au-
have been shown to induce or trigger the autoimmune state. toantibody response that is directly targeted to a single
There is a need to develop and implement appropriate intracellular protein autoantigen. To do so means HgCl2
management and treatment protocols. Animal model studies directly interacts with fibrillarin and/or fibrillarin-containing
help to identify sites for possible drug therapies. snRNPs (the immunogens) by gaining access to the nucle-
The immunoactivating properties of mercury have been olar milieu (Pollard et al., 1997, 2002). This occurs by
grouped into three major pathologic sequelae: lymphopro- cellular toxicity that is nonapoptotically induced by mer-
liferation, hypergammaglobulinemia, and the development cury. Mercury-induced cell death has been closely linked to
of systemic autoimmunity manifested as production of au- the loss of fibrillarin antigenicity and an alteration in the
toantibody and immune complexes (Pollard and Hultman, migratory property of fibrillarin from a 34- to a 32-kDa
1997). Exposure to Hg accelerates autoimmunity in BXSB immunogen under nonreducing sodium dodecyl-Polyacryl-
mice (Guery et al., 1990; Pollard et al., 2001). The results of amide Gel Electrophoresis (SDS-PAGE) conditions (Takeu-
dose–response studies suggest that environmentally relevant chi et al., 1995; Pollard et al., 1999); this indicates that the
tissue levels of mercury are able to exacerbate systemic immunogen is somehow modified to a smaller molecule of
autoimmunity (Pollard et al., 2001). These studies support 32 kDa with a new epitope (Pollard et al., 1999, 2002). The
the concept that low-level (⬍ 40 ␮g of Hg/kg of body modification was presumably caused by mutation of cys-
weight) exposure to mercury can accelerate idiopathic sys- teines to form a disulfide bond in fibrillarin (SOHgOS), a
temic autoimmunity in genetically susceptible hosts. In ad- conformational change that resulted in a neoepitope with a
dition, exposure of BXSB mice to either a short course of different antigenicity. The modified 32-kDa immunogen
high-dose mercury or lower doses over a long period could was found to be a source of T-cell inducers in the process of
trigger systemic autoimmunity. For female BXSB mice, generating self-antibody (Pollard et al., 1999, 2002). The
both the age of initial exposure and the dose of mercury inference is that in the course of metal induced–autoanti-
have been shown to influence the degree of disease exacer- body formation, especially in HgIA situations, different
bation, particularly for antichromatin antibodies (Hultman types of autoantibodies can be produced, some B-cell de-
et al., 1994; Pollard et al., 2001). Studies with MHC- rived and others via T-cell help. In genetically predisposed
identical MRL⫹/⫹ and AKR mice (Pollard et al., 1999) patients, the generation of autoantibodies may be different
suggest that both MHC and non-MHC genes contribute from that generated by nongenetically prone patients. Treat-
significantly to mercury toxicity. MHC and non-MHC ment plans and management schedules therefore are bound
genes also have both been confirmed to contribute to the to vary. More work is needed to help to understand and
expression of idiopathic systemic autoimmunity (Hirsch et elucidate the mechanisms leading to genesis of systemically
ENVIRONMENTAL EXPOSURE TO MERCURY 163

induced autoimmune states as found in the fibrotic pro- data on the mechanisms of such effects are very sparse and
cesses in scleroderma and the development of SLE disease controversial in the available literature (Leonard et al.,
in man. Studies with mercury may assist in probing the 1983b; Ariza et al., 1994). Mutagenic studies with AS52
pathogenesis of these diseases in humans. cells showed that noncytotoxic concentrations (0.1– 0.4
␮M) of Hg2⫹ mercury caused an increase in mutations of
Susceptibility and/or Resistance the gpt gene. These results suggest there may be risks
to Autoimmunity associated with exposure to noncytotoxic levels of Hg2⫹
(Boffetta et al., 1993). Both genotoxic and nongenotoxic
Gene analysis has recently partially revealed the complexity mechanisms probably contribute to the renal carcinogenic-
of regulatory mechanisms of autoimmune susceptibility and ity of mercury (Inouye et al., 1972; Akiyama et al., 2001).
resistance. The actual mechanisms whereby genetically sus- Whitekus and collaborators have shown that in Jurkat cells
ceptible individuals initially enter into the autoimmune cultured in the presence of a CD95 agonist under exposure
phase need to be elucidated. Environmental factors are to noncytotoxic concentrations of HgCl2 (1–10 ␮M), the
known to play a major role in the induction phase of cells continued to proliferate instead of dying (Whitekus et
autoimmunity. Xenobiotics including heavy metals such as al., 1999). Although this is interpreted in light of autoim-
mercury (Mathieson et al., 1992; Takeuchi et al., 1995; Shi mune induction, it also has significance in tumor induction,
et al., 2002), silver (Hultman et al., 1994; Hultman et al., or oncosis. It was established that CD95-mediated apoptosis
1995a), and gold (Pietsch et al., 1989; Schuhmann et al., is attenuated rather than enhanced by inorganic mercury at
1990) are potent environmental immunostimulants that in- the low concentrations specified (ⱕ 10 ␮M) (Whitekus et
duce the production of a number of immunopathologic al., 1999). It appeared that concentration as well as distri-
morbid conditions described earlier: A lymphoproliferation, bution (ie., extracellular vs. intracellular) of Hg2⫹ are quite
hypergammaglobulinemia, and overt systemic autoimmu- important in evaluating the effects of mercury compounds
nity; they also promote the production of antifibrillarin on lymphocyte function. Organomercurials such as MeHg
autoantibodies in H-2s haplotype mice (Hultman et al., are more membrane permeable than Hg2⫹ and are therefore
1995a,b). In mice and other experimental animals the MHC more toxic to lymphocytes (Clarkson, 1997); the mecha-
and non-MHC genes, as well as susceptibility to spontane- nisms for this toxicity might be different from those work-
ous lupus, influence the predisposition to such metal-in- ing to decrease the CD95 death pathway by inorganic mer-
duced immunopathology. Of the various mouse strains, the cury. Bivalent inorganic mercury (Hg2⫹) binds to multiple
DBA/2 uniquely lacks susceptibility to HgIA, despite ex- cell surface receptors via free sulfhydryl groups and results
pressing a susceptible H-2 haplotype (H-2d). In SDF2 inter- in nonspecific receptor clustering, dysregulated signal trans-
crosses between the DBA/2 strain with either SJL or NZB duction, and disorders of cellular function. In fact, the
strain, both of which are highly susceptible to HgIA, a cytotoxicity of Hg2⫹ has been found to increase markedly
single major quantitative trait locus on chromosome 1, the under culture conditions such as supplementation of 2-mer-
Hmr1, has been shown to be common to both crosses. The captoethanol in which the availability of Hg2⫹ to intracel-
region also contains several lupus susceptibility loci. Hmr1 lular targets is facilitated (Whitekus et al., 1999). Therefore,
has been linked to glomerular immune complex deposits, interference with apoptosis by mercury supports findings
not to autoantibody production (Morel et al., 1994; Drake et showing that autoimmune diseases are often caused by a
al., 1995; Hogarth et al., 1998; Kono et al., 2001). This failure to delete autoreactive lymphocytes. Obviously, if
suggests that the DBA/2 strain’s resistance to HgIA proba- lymphocytes accumulate as lymphomas, then there is a link
bly involves primarily the later genes of disease pathogen- to carcinogenesis. Results of recent studies in our laboratory
esis. Research indicates that DBA/2 resistance appears to be show that mercury induces cytotoxicity and transcription-
multigenic and largely a result of the effects of the Hmr1 ally activates a significant number of stress genes in human
gene and, to a lesser extent, a locus on chromosome 7 (Kono liver carcinoma cells, including those involved in cell cycle
et al., 2001). Interestingly, the DBA/2 strain is also resistant regulation and apoptosis (Sutton et al., 2002; Tchounwou et
to both silver- and gold-induced autoimmunities (Schuh- al., 2002).
mann et al., 1990; Hultman et al., 1995b). These findings Heavy metals such as lead and mercury, organic sol-
indicate a probable relationship of Hmr1 resistance to mer- vents, alcohol, and ionizing radiation are confirmed envi-
cury-induced disease to broad xenobiotic agents. ronmental teratogens (Harris et al., 1973). Animal studies
have shown that low levels of mercuric chloride or phenyl
mercuric acetate enter fetal tissues and result in abortion,
GENOTOXIC, CARCINOGENIC, AND growth retardation, subcutaneous edema, encephaly, and
TERATOGENIC EFFECTS OF MERCURY anophthalmia (Nierenberg et al., 1998). Teratogenic effects,
such as cleft lip and palate, rib fusions, and syndactylies
Mercury is a highly deleterious environmental pollutant have been seen. Methylmercury compounds are known to
with recognized mutagenic and teratogenic effects, although be teratogenic in multiple organs in rats, hamsters, and mice
164 TCHOUNWOU ET AL.

(Harris et al., 1973; Myers et al., 1995). Skeletal malforma- retention of metallic mercury may facilitate the conversion
tions, rib defects, abnormal skeletal calcification, cleft lip of metallic, elemental mercury to divalent mercury and its
and palate, micrognathia, and clubfeet have all been seen subsequent absorption, with the development of hepatic
after mercury intoxication during the fetal period. In non- dysfunction (Lin and Lim, 1993). Most MeHg is eliminated
pregnant rats methylmercury accumulates in nerve cells from the body by demethylation and by excretion of the
(neuroglia) and nerve fibers. The dorsal root ganglia are inorganic form in the feces. The processes of biliary secre-
well invaded, whereas the Purkinje cells of the cerebellum, tion and demethylation by microflora do not occur in suck-
ventral horn motor cells, and cerebellar granule cells show ling animals. The role of these two processes in suckling
fewer invasions. In contrast, mercuric chloride intoxication human infants is unknown.
is highest in the dorsal root ganglia, followed by the ventral Nutritional factors, not the primary absorption site,
horn motor neurons, calcarine cortical neurons, and the vastly influence the reabsorption rate of MeHg (Yannai and
granule cells of the cerebellum (Chan, 1998). Sachs, 1993); and a vast variety of foods and nutrients alter
MeHg metabolism. The alkalinity of the environment has
been associated with increased absorption of mercury in the
DIETARY INFLUENCES ON MERCURY form of Hg2⫹ ions. Milk may also promote the reabsorption
TOXICITY AND COUNTEREFFECTS of MeHg after enterohepatic circulation (Endo et al., 1984).
ON DIET It is likely that diet affects the absorption of organic and
inorganic Hg (Nielsen and Andersen, 1995) via combina-
Epidemiological data on the routes of Hg exposure in hu- tions of different mechanisms. Nutrients do interact with the
man populations indicate that the major sources of MeHg metabolism of MeHg at the physiologic level. Nutrients
exposure arise from fish and marine products (Davidson et play important roles in the bioavailability, toxicity, and
al., 1998). It is estimated that fish and fish products contrib- dissemination of molecules throughout the organs that are
ute a range of 20%– 85% of the MeHg intoxication in susceptible. They also influence the biochemical, immuno-
various populations. Drinking water, cereals, vegetables, logic, or cytotoxic responses to Hg. Data are lacking, how-
and meat are believed to make important contributions to ever, on the mechanism of MeHg toxicity; nutritional mod-
MeHg burden in man (Galal-Gorchev, 1993). Dental amal- ifiers of mercury toxicity are not well studied. Nutritional
gams have been more or less implicated in the release of Hg factors, including selenium (Mykkanen and Metsanitty,
vapor (Roberts et al., 2001). The general consensus is that 1987; Magos, 1991), vitamin C (Solomons and Viteri,
the daily intake of MeHg comes from its concentration in 1982), vitamin B (Levander and Cheng, 1980; Bender,
foodstuffs and that the naturally present inorganic Hg in the 1984), and essential minerals (Levander and Cheng, 1980;
hydrosphere and biosphere because of acid rain and indus- Chowdhury and Chandra, 1987; Zorn and Smith, 1990;
trial mining activities and the follow-up biomethylation of Lugea et al., 1994) have also been implicated in the modi-
this Hg is a risk for global Hg exposure, which is on the fication of MeHg toxicity. However there is not general
increase (Fitzgerald and Clarkson, 1991). These reports agreement on the role these supplements play in the toxicity
mean that for any population the mechanism of total Hg of MeHg. No protective or antagonistic effects of dietary
intake and its effects may be multifactorial and probably supplements on the toxicity of MeHg to humans have been
relies heavily on dietary habits for that population (Chap- clearly established.
man and Chan, 2000). The duration and timing of exposure Experimental data on animal models have been the main
to MeHg have been found to be significant critical factors as sources regarding the role of nutrients on MeHg intoxica-
well. For instance, the effects of prenatal exposure have tion. Ethanol is generally known to have a synergistic effect
been determined to be more important than the effects of in the toxicity of MeHg. Mortality from MeHg intoxication
exposure via breast-feeding in mice (Nielsen and Andersen, with superimposed ethanol action is known. Ethanol pro-
1995). The role of nutrition on MeHg toxicity has generally motes kidney pathogenesis by decreasing the activities of
been approached from two areas: effects of nutrients on Hg amino acid transferases and creatine phosphokinases (Rum-
metabolism and vice versa (Galal-Gorchev, 1993). beiha et al., 1992). Selenium absorption is reduced in Se–Hg
Methylmercury is absorbed throughout the intestine; up interactions (Mykkanen and Metsanitty, 1987), suggesting
to 90% of MeHg crosses most biological membranes (Nors- that Hg reduces the transfer of selenite from the intestine to
eth, 1970). It binds in vivo to proteins such as albumin and the body but not the transfer of selenium-methionine (Se-
sulfur-containing proteins (Yasutake et al., 1990; Adachi et Met). Rather, MeHg disrupts nutrient transport such as the
al., 1994). In adults recycling occurs in the enterohepatic exchange of Met and Se through the BBB (Aschner and
system (Norseth, 1970). It is secreted into bile and partly Aschner, 1990). Fetal uptake of nutrients such as Se, vita-
reabsorbed into portal circulation and thereby returned to min B12, and Zn in mice (Danielsson et al., 1984) and
the liver. A fraction of biliary mercury is converted by humans (Lin and Lim, 1993) is altered by mercuric ions
microflora to inorganic mercury, which is reabsorbed only (Hg2⫹) during pregnancy. The binding of Hg2⫹ to trans-
to a small extent (ATSDR, 1999). Massive and prolonged membrane thiol groups has been implicated in the decline of
ENVIRONMENTAL EXPOSURE TO MERCURY 165

the sugar-Na⫹ phlorizin-sensitive cotransport system, lead- suggested that Se modifies MeHg so that the latter is re-
ing to the inhibition of galactose absorption in rats (Fujinka leased from blood proteins (Magos et al., 1984). Zinc has
and Zasshi, 1987); cysteine reversed this interaction. Cys- the effect of reducing lipid peroxidation by enhancing the
teine is believed to act synergistically with MeHg neuro- activities of enzymes such as GSH peroxidase to repress
toxicity (Lugea et al., 1994). Methylmercury is transported mercury-induced neurotoxicity (Gale, 1984; Fukino et al.,
in the circulation bound to serum proteins such as albumin 1986). Iron, on the other hand, is noted for reducing MeHg
and mercaptoalbumin and enters major organs like the kid- toxicity; dexoferoxamine, an iron chelator, has been found
neys, liver, and brain (Mokrzan et al., 1995) as well as the to inhibit the MeHg-induced formation of excess reactive
placenta and fetus in pregnancy (Davidson et al., 1998). Red oxygen species in rats’ brains (LeBel et al., 1992).
blood cells and epithelial tissues are also invaded in signif-
icant amounts (Norseth and Clarkson, 1970).
L-Leucine, L-methionine, and 2-amino,2,norborane car-
Vitamins
boxylic acid may inhibit the uptake of MeHg through the
amino acid transport system (Mokrzan et al., 1995). MeHg Vitamins E and C have antioxidant properties and are ex-
enters the brain in the form of an L-Cys complex by the pected to negate the formation of active oxygen species
amino acid transport system (Aschner and Aschner, 1990). produced in the course of MeHg metabolism (Chang et al.,
Cystine has been implicated as one nutrient that may in- 1978). Vitamin E protects against ataxia, paralysis of the
crease MeHg neurotoxicity, although it can enter other hind limbs, and necrosis in the brains of rats and hamsters
organs by any one of several transport systems, including (El-Begearmi et al., 1976; Chang et al., 1978); it protects
the facilitated D-glucose and the Cl⫺ ion transport system animals against the effects of organic mercury toxicity but
(Norseth and Clarkson, 1970). The association of MeHg not against Hg2⫹ toxicity (Chang et al., 1978), and the
with small-molecular-weight thiol compounds enhances the protection afforded to animals extends from parent to off-
transport of MeHg both into and out of cells (Mokrzan et al., spring (El-Begearmi et al., 1976; Chang et al., 1978). The
1995). In general, diet composition has been shown to affect role of vitamin C in mercury toxicity is contradictory at
the distribution and toxicity of MeHg (Hojbjerg et al., 1992; best. The recovery of enzyme (alpha- and beta-galactosi-
Adachi et al., 1994; Hojbjerg, 1996). The percentage of dase) activity after vitamin C treatment of MeHgCl-exposed
fiber in the diet affects the retention of Hg; pectin, wheat mice was found to be incomplete and seemed to be organ
bran, and cellulose can alter the ability of microflora to dependent (Vijayalakshmi et al., 1992). There were indica-
demethylate MeHg and therefore affect its reabsorption rate. tions that although susceptible organs such as the kidneys or
For example, wheat bran enhances fecal excretion of Hg the liver may be relieved of Hg burden, neurotoxicity in
after MeHg exposure by increasing the demethylation rate other organs such as the brain is enhanced. Vitamin A was
of MeHg in intestinal flora. Sufficient protein intake pro- not found protective against in vivo treatment of MeHg
longs survival after oral MeHg dosing (Rowland et al., toxicity; rather, it enhanced MeHg toxicity in in vivo studies
1986). with rats (Bapu et al., 1994).

Minerals Combined Nutrient Effects


Selenium (Se) plays a protective role against MeHg toxicity Research on Hg2⫹ methylation found that simultaneous
(Mykkanen and Metsanitty, 1987; Magos, 1991). However, exposure to folate, vitamin B12, and ascorbate increases
fish and marine mammals, the main sources of Hg in the MeHg in the liver and in hair, whereas the combination of
diet, also contain a rich source of Se (Mykkanen and Met- vitamin C and vitamin B12 increases MeHg in the brain
sanitty, 1987; Cuvin-Aralar and Fumess, 1991). Epidemiol- (Welsh, 1977). Vitamin E and Se seem to have an additive
ogists therefore consider Se an important but unproven antioxidant interaction. At low Se concentrations the two
modifier of MeHg intoxication (Tan and Perkin, 2000). combinations afford synergistic protection against MeHg
Sufficient intake of Se and Zn may protect against MeHg (Bender, 1984; Welsh and Soares, 1975). These findings are
toxicity. Selenium, in particular, is found to counteract the now used generally in the management of Hg toxicity.
toxicity of other heavy metals including cadmium, thallium, Doses of 400 IU/day of vitamin E have been shown to have
and silver (Mykkanen and Metsanitty, 1987). Selenium is a protective effect when the brain is exposed to methylmer-
believed to delay rather than prevent intoxication from cury (Chang et al., 1978; King and Soares, 1981). Doses of
MeHg (Magos et al., 1984). The mechanisms for delaying 200 – 400 ng/day of Se are utilized particularly for mercury
both inorganic and organic mercury intoxication implicate elimination in the most intoxicated patients to facilitate the
the formation of Hg–Se complexes, GSH–Se–Hg and bis function of glutathione in mercury detoxification (Levander
selenide (methylmercuric selenide), respectively (Myk- and Cheng, 1980; King and Soares, 1981; Hirsch et al.,
kanen and Metsanitty, 1987; Magos, 1991). It has been 1982; Mykkanen and Metsanitty, 1987).
166 TCHOUNWOU ET AL.

Mercury inhibits protein synthesis in general by inacti- tests. Follow-up studies of workers previously exposed to
vating enzymes such as aspartate and alanine amino acid high levels of mercury vapor but not exposed for 10 or more
transferases, although MeHg appears to stimulate protein years prior to being examined revealed that adverse effects
synthesis in the brain (Solomons and Viteri, 1982; may persist on the nervous system. Mathiesen and col-
Kuznetsov, 1987). Mercury exposure is found to alter the leagues examined 70 workers who had been exposed for a
lipid profiles including fatty acid and cholesterol production period of 1–35 years, with a mean of 12.7 years. The
(Chowdhury and Chandra, 1987; Bano and Hasan, 1989). average yearly exposure was measured as 8 –584 ␮g Hg/m3
Triglyceride levels declined in the central nervous system of (ATSDR, 1999; Dewailly, 1997). Compared to the perfor-
MeHg-exposed rats, whereas tocopherol levels increased in mance in a control group, these workers showed decreased
serum (Chowdhury and Chandra, 1987). Many enzymes in performance in a number of neuropsychologic attributes.
carbohydrate metabolism, including glucose-6-phosphatase, Despite these high exposure levels, no residual effects were
amylase, maltase, and lactase, are inhibited by exposure to observed on general intellectual ability or ability to reason
Hg2⫹ (Rana and Sharma, 1982). Calcium channel blockers logically.
(CCBs) alleviate the toxic effects of MeHg on the serum Epidemiological evidence derived from studies of the
electrolytes sodium and potassium via their blocking action effects of the environment on the toxic susceptibility of
(Chowdhury and Chandra, 1987). Sakamoto and colleagues different communities to MeHg has come from the Amazon
determined that CCBs prevented a decline in body weight (Lodenius and Malm, 1998), the Seychelles Republic
and neurologic symptoms in rats (Sakamoto et al., 1996). (Marsh et al., 1995), and the Faroe Islands (Clarkson, 1991;
De Flora et al., 1994). Through the consumption of fish and
other seafood, many populations have been exposed to
equivalent doses of MeHg (Sweet, 2001). Some populations
MERCURY RISK ASSESSMENT AND
showed subsequent neurotoxic effects, and others did not
MANAGEMENT
(Gilbert and Grant-Webster, 1995). This observation has
prompted a search for the role of nutritional status of vari-
Minimum Risk Levels
ous populations in combating or evading drastic toxicolog-
Currently, health authorities are concerned with the risk ical effects of MeHg intoxication (Chapman and Chan,
associated with mercury exposure. Because of the possible 2000). Up to now there has been no clear data as to the best
teratogenic effects of mercury on humans, health authorities in vivo nutritional modifier of MeHg toxicity. However, it is
are faced with the decision of deriving the highest possible worth noting that MeHg attains its highest concentrations in
level of oral mercury exposure, the reference dose (RfD), the tissues of long-lived edible predatory fish (Inouye et al.,
the level considered to present no risk to the public in 1972). It is a toxic compound that is well absorbed from the
general and to pregnant women in particular. Using an diet despite having no demonstrable biologic requirement in
excretion rate of 1% per day, it was computed that for a humans (Sweet, 2001). This diet has been presumed to be
70-kg person a daily Hg intake of 0.3 mg/day would yield the main source of exposure in human populations
a blood value of 0.2 ␮g/mL, corresponding to the lowest (ATSDR, 1999). Daily intake of MeHg seems to depend to
blood level known to be toxic (ATSDR, 1999). The RfD can a large extent on its concentration in foodstuffs and on the
be taken as the dose that can be ingested daily for a lifetime dietary habits of the consumer.
without a significant risk of adverse effects. With a safety Currently, the Food and Agriculture Organization (FAO)
factor of 10, the daily intake of MeHg was set at 0.03 and the World Health Organization (WHO) have set levels
mg/day for a 70-kg person, corresponding to 0.025 mg/day considered a tolerable intake as 3.3 ␮g/kg/week or 200
for a 60-kg woman. For pregnant women the suggestion is ␮g/week of methylmercury based on the level at which
not to consume fish containing greater than an arbitrary paresthesia presents in adults (Chapman and Chan, 2000).
MeHg concentration of 0.25 ␮g/g and that women of child- But the fetus is particularly sensitive to MeHg, even at a
bearing age should not work in areas at risk to MeHg (alkyl) concentration that results in few or no clinical signs of
exposure (ATSDR, 1999). maternal toxicity (Sweet, 2001). High levels of prenatal
In general, air concentrations in the workplace above 50 MeHg exposure can induce cerebral palsy, mental retarda-
␮g Hg/m3, corresponding to steady-state urinary excretion tion, low birth weight, and early sensorimotor dysfunction
rates of creatinine of 60 ␮g Hg/g are associated with fine (Gilbert and Grant-Webster, 1995). This has prompted a
tremors in the extremities that frequently are not noticed by further review of reference doses for MeHg in view of its
the worker (Kulig, 1998). Slowed nerve conduction velocity prenatal developmental effects, infant exposure, and the
is another preclinical effect found at these lower levels. important objective of establishing the highest level that can
Studies on dentists have suggested adverse effects at air be considered tolerable for human exposure (Inouye et al.,
concentrations lower than 50 ␮g Hg/m3 (U.S. EPA, 1997). 1972; Stem, 1993; Chapman and Chan, 2000).
Average air concentrations as low as 14 ␮g Hg/m3 were Many regulatory agencies have utilized findings from
associated with decreased performance on psychomotor two large longitudinal studies of effects of prenatal Hg
ENVIRONMENTAL EXPOSURE TO MERCURY 167

exposures from seafood consumption on child neurodevel- have proved negative (Philbert et al., 2000). It is now
opment; these studies were conducted in the Republic of believed that a time-integrated assessment of absorption
Seychelles, where 85% of the population lives on ocean fish may be a determining factor for toxic neuropathies resulting
(De Flora et al., 1994; Davidson et al., 1998), and in the from prolonged low-level exposures. Therefore, the level of
Faroe Islands (Cernichiari et al., 1995; Clarkson, 1991; Hg detected in the hair is now believed to be a more
Davidson et al., 1998). The Seychelles studies reported appropriate but unproven alternative for monitoring Hg
mean maternal hair total Hg level of 6.8 ppm and mean toxicity (Cernichiari et al., 1995).
child hair total Hg level at 66 months of age of 6.5 ppm In pregnancy this presents another level of complication.
(Davidson et al., 1998). No adverse pathology at 66 months No data clearly present the effect of mercury on exposed
was correlated with prenatal or postnatal MeHg exposure. pregnant women except retrospective studies arising from
The second study, of 1022 singleton births, revealed that at disasters (Amin-Zaki et al., 1981). Estimates of reasonably
approximately 7 years of age some of the children in the safe exposure have been based on pharmacological princi-
study still showed no Hg-related damage after exhaustive ples (Clarkson, 1997; Nierenberg et al., 1998; Philbert et al.,
clinical neurophysiological tests (Marsh et al., 1995; Myers 2000); taking into account the amount ingested, duration of
et al., 1995; Davidson et al., 1998). However, subsamples of exposure, excretory rate, and sensitivity. A certain mercury
112 and 217 children whose mothers had hair Hg concen- dose delivered acutely may or may not present with toxic
trations of 10 –20 ppm and 7.1 ppm, respectively, were effects, and the same amount distributed over a period of
compared to a subsample of children whose mothers had time may still give no evidence of mercury poisoning. Yet
hair mercury below 3 ppm. Mild decrements were noted in there is no certainty that the same dose will present the same
the former samples, especially in the domains of motor signs and symptoms in two different individuals. Methyl-
function, language, and memory (Davidson et al., 1998). mercury accumulates in the body at 100 times the daily
Based on such findings, Health Canada revised the tolerable intake at equilibrium (WHO, 1990), and individual varia-
limit of daily Hg intake for women of childbearing age and tions in sensitivity to mercury compounds have been ob-
infants to 0.2 ␮g/kg of body weight/day (Galal-Gorchev, served from epidemiological studies. In Iraq, the half-life of
1993). The U.S. Environmental Protection Agency (EPA) methylmercury elimination was found to vary from 40 to
set the reference dose for MeHg at 0.1 ␮g/kg of body 105 days, with a mean of 65 days (Bakir et al., 1973;
weight/day (Stem, 1993; U.S. EPA, 1997; Rice et al., 1999). Amin-Zaki et al., 1974). The body burden at which pares-
The nervous system seems to be most affected in mer- thesia in adults could be detected in Iraq varied from 25 to
cury intoxications (Philbert et al., 2000). Because of the 200 mg. The chemical form of mercury also determines its
unique structural and functional characteristics of the ner- toxicity. In mice given intraperitoneal injections acute LD50
vous system, neurotoxicity has features distinct from the values for mercuric chloride, phenylmercury, and methyl-
toxic effects in other organs. Neurotoxicants show selective mercury have been reported to be 5, 8, and 14 mg of Hg/kg
toxicity for cells or areas of the CNS and peripheral nervous of body weight, respectively (Clarkson, 1997; Nierenberg et
system, but the mechanisms underlying such selectivity are al., 1998). Inorganic mercury appeared to be the most toxic
relatively evasive. The relationship among the biochemical, in acute administration; in humans, however, chronic oral
anatomical, and physiological parameters within the ner- mercury intake resulting in minimal toxic effects has been
vous system are not fully understood. It is therefore difficult estimated to be 28 mg/day for inorganic salt Hg2⫹, 0.75
to evaluate certain biochemical modifications observed in mg/day for phenylmercury, and 0.3 mg/day for methylmer-
animals or humans following exposure to neurotoxicants. cury (Philbert et al., 2000).
Establishing a correlation between mercury level in humans Apparently there is no clear-cut evidence supporting any
and toxic reaction in the nervous system has been attempted of the safe limits set by the regulatory agencies. Besides, it
in various studies (Clarkson, 1991; Stem, 1993; Gilbert and is becoming quite clear that a low level of exposure to
Grant-Webster, 1995; Marsh et al., 1995; U.S. EPA, 1997; mercury may be hazardous to the immune system, as dis-
Rice et al., 1999; Philbert et al., 2000). Most extrapolations cussed earlier. Based on the currently available scientific
are made from animal studies, and the findings may or may information on the adverse health effects of mercury, it is
not reflect a true state in humans. probably safe to say that zero tolerance of mercury may be
In humans functional and pathological nervous tissue the objective for most health authorities and policy makers.
alterations induced by exogenous chemicals can also be
caused by several other unrelated factors including stress,
social environment, and lifestyle. The complexity of these
Diagnosis of Mercury Intoxication
factors makes the search for biological indicators of neuro-
toxicity a huge undertaking. The most common approach, of Mercury intoxication has been diagnosed primarily based
searching for a correlation between Hg concentration in on history of exposure and symptoms of intoxication. Acute
blood or urine and the appearance of symptoms or electro- exposure to mercury vapor can cause coughing, dyspnea,
neurographic changes, may not be the best one; most results chest pain, cyanosis, excitement, and tremor (Albers et al.,
168 TCHOUNWOU ET AL.

1988). Chronic exposure may lead to subtle symptoms such and generally can be grouped into natural processes and
as weakness, fatigue, anorexia, weight loss, and gastroin- sources originating from human activities. Geogenic pollu-
testinal disturbances (Amin-Zaki et al., 1974; Dewailly, tion by mercury arises from the earth’s crust and oceans,
1997; Prati et al., 2002). Inorganic mercury ingestion in- erosion of mineral deposits, and volcanoes. Human activi-
duces symptoms of acute thirst, metallic taste, nausea, and ties such as metal smelting, coal production, chemical syn-
abdominal pain, followed by tenesmus, bloody diarrhea, thesis and use, and waste disposal are means of adding large
stomatitis, gastritis, colitis, renal tubule degeneration, and quantities of mercury to the environment. Interconversions
death (Choi et al., 1978; Albers et al., 1988; Dewailly, of mercurous species by microorganisms lead to bioaccu-
1997). Chronic ingestion results in excessive salivation, mulation of mercury. Biomethylation and concentration of
loose teeth, and gingivitis. Neurologic signs include irrita- mercury in bodies of water play significant roles in the
bility, nervousness, tremors, and slurred speech. Paresthesia spread of mercury throughout the environment.
occurs in the distal extremities with methylmercury intoxi- Problems arising from mercury contamination are en-
cation, followed by fatigue, headache, and neurasthenia. hanced when mercury gets into the food chain. Throughout
There is a prolonged interval between exposures to meth- the world exposure to mercury occurs via ingestion, inha-
ylmercury, in particular, and the appearance of symptoms lation, dental amalgams, and dermal contact; however, the
that last up to several weeks in some cases (Bakir et al., food chain seems to be the predominant route of human
1973; Amin-Zaki et al., 1974). exposure to methylmercury, which is the most toxic form of
mercury. These avenues of pollution present great chal-
lenges to public health. Mass outbreaks of mercury poison-
Treatment Schedules
ing in many parts of the world including China, Japan, Iraq,
A major challenge is the treatment of pregnant women the former Soviet Union (now Russia), the Seychelles, the
exposed to mercury. Ideally, prevention from any further Faroe Islands and Ghana have prompted health profession-
contact with the source is the first step in management. als to take a second look at the public health impact of
Supportive care is given when necessary to maintain vital mercury toxicity. After the largest outbreak, which occurred
functions. More definitive treatment is based on knowing in rural Iraq during the winter of 1971–72, as many as
the type and degree of exposure. In treatment of acute 40,000 individuals were found to suffer from mercury poi-
ingestion of inorganic Hg, dosing with egg whites or milk is soning resulting from consuming homemade bread prepared
recommended to decrease further absorption of mercuric from mercury-contaminated seed grains. In most cases se-
ions (Albers et al., 1988). A 5% sodium formaldehyde vere brain damage was detected from prenatal exposure.
sulfoxylate solution by mouth decreases mercury absorp- These children were found to have difficulties in achieving
tion. The use of dimercaprol (BAL), calcium disodium developmental milestones at the appropriate time.
edetate, and N-acetyl-D, L-penicillamine assists the body’s Amalgam fillings have also been associated with a vari-
ability to eliminate mercury from the tissues. BAL is useful ety of problems such as Alzheimer’s disease, infertility,
for severe systemic poisoning; it combines with mercury food allergies, multiple sclerosis, and thyroid and kidney
and forms a complex excreted by the kidneys (El-Begearmi dysfunction. Organic mercury compounds are generally
et al., 1976; King and Soares, 1981). BAL use is not more toxic, with their toxicity centered in the CNS, though
recommended for chronic exposure, however, because it the kidneys and the immune system may also be signifi-
enhances brain uptake of mercury and has local as well as cantly affected. Prenatal and postnatal exposure to mercury
systemic side effects. Penicillamine also reacts reversibly adversely affects the CNS, but toxicity is pronounced from
with mercury and is able to break mercury/protein com- exposure during the prenatal period, when neurogenesis is at
plexes (Philbert et al., 2000). Methylmercury poisoning is its height. In adults exposure to mercury can result in visual
treated symptomatically. Chelators are of limited use be- and hearing impairment, tremor, and muscle spasticity,
cause of adverse side effects. Pyridoxine-5-thiol has been leading to coma and death. One of the most serious threats
shown to reduce MeHg concentration in the brain and liver from mercury toxicity, however, is insidious, involving the
of poisoned rats; thiol resins effectively increase MeHg development of an autoimmune state in genetically predis-
excretion, whereas polythiol resin as 1% concentrate with posed individuals. Although mercury is capable of inducing
food increases MeHg excretion in mice (Vijayalakshmi et an immunosuppressive state with continuous exposure, it
al., 1992). also has the ability to cause defects in programmed cell
death that eventually end in the generation of lupuslike
symptoms and possibly carcinogenesis. Mercury toxicity
CONCLUSIONS depends on exposure route, frequency, dose level, nutri-
tional status, individual susceptibility, and genetic predis-
Mercury is one of the toxic elements that are widely dis- position. Many guidelines have been developed to protect
tributed in nature. Available scientific data show that the public health; however, the mercury reference dose needs to
sources of mercury polluting the environment are numerous be revisited in light of recent findings indicating that low
ENVIRONMENTAL EXPOSURE TO MERCURY 169

levels, as little as 0.1– 0.4 ␮M of mercury, induce mutations Aschner M, Lorscheider FL, Cowan KS, Conklin DR, Vimy MJ,
in cultured cells. Understanding the general risk involved in Lash LH. 1997. Metallothionein induction in fetal rat brain and
mercury pollution and toxicity is a prerequisite for devel- neonatal primary astrocyte cultures by in utero exposure to
oping a useful health advisory document for mercury com- elemental mercury vapor (Hg0). Brain Res 778:222–232.
pounds. Therefore, it is important to know the physical and Ashkenazi A, Dixit VM. Death receptors: signaling and modula-
chemical properties, sources and pathways of exposure, tion. 1998. Science 281:1305–1317.
systemic and carcinogenic health effects, current regulatory Ashkenazi A, Dixit VM. 2002. Death receptors: Signaling and
and health guidelines, and diagnostic and treatment sched- modulation. Science 281(5381):1305–1326.
ules that can assist in developing a comprehensive risk Ask K, Äkesson A, Berglünd M, Vahter M. 2002. Inorganic
assessment and in implementing appropriate risk manage- mercury and methylmercury in placenta of Swedish Women.
ment strategies for mercury. Env Health Perspect 110(5):523–531.
Aten J, Bossman CB, Rozing J, Stijnen T, Hoedenmaeker PJ,
Weening JJ. 1998. Mercuric chloride-induced autoimmunity in
the brown Norway rat; cellular kinetics and major histocompat-
REFERENCES ibility complex antigen expression. Am J Pathol 133:127–134.
Abbas AK, Murphy KM, Sher A. 1996. Functional diversity of Agency for Toxic Substances and Disease Registry [ATSDR].
helper T lymphocytes. Nature 383:787–796. 1999. Toxicological profile for mercury: TP-93/10. Atlanta,
Georgia: Centers for Disease Control.
Adachi T, Yasutake A, Hirayama K. 1994. Influence of dietary
protein and sulfur amino acids on the fate of methyl mercury in Bakir F, Damluji S, Amin-Zaki L, Murtadha M, Khalidi A, Al-
mice. Toxicology 93:225–234. Rawi N, Tikriti S, Dhahir H, Clarkson T, Smith J, Doherty R.
1973. Methylmercury poisoning in Iraq. An interuniversity re-
Adams CR, Ziegler DK, Lin J. 1983. Mercury intoxication simu-
port. Science 181:230 –243.
lating amyotropic lateral sclerosis. J Am Med Assoc 250:642–
643. Bano Y, Hasan M. 1989. Mercury induced time-dependent alter-
ations in lipid profiles and lipid peroxidation in different body
Agocs MM, Etzel RA, Parrish RG, Paschal DC, Campagna PR,
organs of cat-fish Heteropneustes fossilis. J Environ Sci Health
Cohen DS, Kilbourne EM, Hesse JL. 1990. Mercury exposure
8(24):145–166.
from interior latex paint. N Engl J Med 323:1096 –1101.
Bapu C, Vijaylakshmi K, Sood PP. 1994. Comparison of mono-
Akiyama M, Oshima H, Nakamura M. 2001. Genotoxicity of
thiols and vitamin therapy administered alone or in combina-
mercury used in chromosome aberration tests. Toxicol in Vitro
tions during methyl mercury poisoning. Bull Environ Contam
15(4 –5):463– 477.
Toxicol 52:182–189.
Albers JW, Kalenbach LR, Fine LJ, Lengulf GD, Wolfe RA,
Bargagli R, Iosco FP, Barghigiani C. 1987. Assessment of mercury
Donofrio PD, Alessi AG, Stolp-Smith KA, Bromberg MB.
dispersal in an abandoned mining area by soil and lichen anal-
1988. Neurological abnormalities associated with remote occu-
ysis. Water Air Soil Pollut 36:219 –225.
pational elementary mercury exposure. Anal Neurol 24:651–
659. Barghigiani C, Bargagli R, Siegel BZ, Siegel SM. 1990. A com-
parative study of mercury distribution on the Aeolian volcanoes;
Amin-Zaki L, Elhassani S, Majeed MA, Clarkson TW, Doherty
volcano and stromboli. Water Air Soil Pollut 53:179 –188.
RA, Greenwood M. 1974. Intra-uterine methyl mercury poison-
ing in Iraq. Pediatrics 54(5):587–595. Bariety J, Druet P, Laliberte F, Sapin C. 1971. Glomerulonephritis
Amin-Zaki L, Majeed MA, Greenwood MR, Elhassani SB, Clark- with ␥- and 1C-globulin deposits induced in rats by mercuric
son TW, Doherty RA. 1981. Methylmercury poisoning in Iraqi chloride. Am J Pathol 65:293–301.
suckling infant: a longitudinal study over five years. J Appl Bell LK, Ball R, Pratt RD. 2001. An assessment of thimerosal use
Toxicol 1:210 –214. in childhood vaccines. Pediatrics 107:1147–1154.
Andre I, Gonzalez A, Wang B, Katz J, Benoist C, Mathis D. 1996. Bender DA. 1984. B-vitamins in the nervous system. Neurochem
Checkpoints in the progression of autoimmune disease: lessons Int 6:297–321.
from diabetes models. Proc Natl Acad Sci USA 93:2260 –2274. Berden JH. 1997. Immunology in medical practice. III. Dissemi-
Ariza ME, Holliday J, Williams MV. 1994. Mutagenic effect of nated lupus erythematosus: disturbed apoptosis? Ned Tijdschr
mercury (II) in eukaryotic cells. Toxicol in Vivo 8(4):559 –563. Geneeskd, 39:1848 –1854. (Dut).
Arnett FC Jr. 1993. The genetic basis of lupus erythematosus. In: Biancone L, Andres G, Ahn H, Lim A, Dai C, Noelle R, Yagita H,
Wallace D, Hahn BH, editors. Dubois’ lupus erythematosus, 4th DeMartino C, Stamenkovic I. 1996. Distinct regulatory roles of
ed. Philadelphia: Lea & Ferbiger. p 13–36. lymphocyte costimulatory pathways on T helper type 2-medi-
Arnett FC, Reveille JD, Goldstein R, Pollard KM, Leaird K, Smith ated autoimmune disease. J Exp Med 183:1473–1481.
EA, LeRoy EC, Fritzler MJ. 1996. Autoantibodies to fibrillarin Bittner AC Jr, Echeverria D, Woods JS, Aposhian V, Naleway C,
in systemic sclerosis (scleroderma): an immunogenetic, serolog- Martin MD, Mahurin RK, Heyer NJ, Cianciola M. 1998. Be-
ical and clinical analysis. Arthritis Rheum 39:1151–1162. havioral effects of low-level exposure to Hg0 among dental
Aschner M, Aschner JL. 1990. Mercury neurotoxicity: mecha- professionals: A cross-study evaluation of psychomotor effects.
nisms of blood– brain barrier transport. Neurosci Biobehav Rev Neurotoxicol Teratol 20(4):429 – 439.
14:169 –176. Boadi WY, Shurtz-Swirski R, Barnea ER, Urbach J, Brandes JM,
170 TCHOUNWOU ET AL.

Yannai S. 1992. The influence of mercury on the secretion of Clarkson TW. 2002. The three modern faces of mercury. Environ
human chorionic gonadotropin in superfused young placenta Health Perspect 110(1):11–24.
tissue. Pharmacol Toxicol 71:19 –23. Cleary D. 1990. Anatomy of the Amazon gold rush. Iowa City, IA:
Boffetta P, Merler E, Vainio H. 1993. Carcinogenicity of mercury University of Iowa Press.
and mercury compounds. Scand J Work Environ Health 9(1): Cuvin-Aralar MLA, Fumess RW. 1991. Mercury and selenium
1–7. interaction: a review. Ecotoxicol Environ Saf 21:348 –364.
Bolland S, Ravetch JV. 2000. Spontaneous autoimmune disease in Dales L, Kahn E, Wei E. 1971. Methylmercury poisoning. An
Fc(␥)RIIB-deficient mice results strain-specific epistasis. Immu- assessment of the sportfish hazard in California. Calif Med
nity 13:277–289. 114(3):13–15.
Bottomly K, Constant SI. 1997. Induction of Th1 and Th2 CD4⫹T Danielsson BR, Dencker L, Khayat A, Orsen I. 1984. Fetotoxicity
cell responses: the alternative approaches. Annual Rev Immunol of inorganic mercury in the mouse: distribution and effects on
15:297–311. nutrient uptake by placenta and fetus. Biol Res Preg Perinatol
Casiano CA, Martin SJ, Green D, Tan EM. 1996. Selective cleav- 5:102–109.
age of nuclear autoantigens during CD95 (Fas/APO-1)-medi- Davidson PW, Myers GJ, Cox C, Axtell C, Shamlaye C, Sloane-
ated T cell apoptosis. J Exp Med 184:765–771. Reeves J, Cermichiari E, Needham L, Choi A, Wand YN, Berlin
Casiola-Rosen LA, Anhalt GJ, Rosen A. 1995. DNA-dependent M, Clarkson TW. 1998. Effects of prenatal and postnatal meth-
protein kinase is one of a subset of autoantigens specifically ylmercury exposure from fish consumption on neurodevelop-
cleaved early during apoptosis. J Exp Med 182:1625–1632. ment. Outcomes at 66 months of age in the Seychelles Child
Development Study. J Am Med Assoc 280(8):701–707.
[CDC/AAFP/AAP/ACIP] Centers for Disease Control and Preven-
Deapen D, Escalante A, Weinrib L, Horwitz D, Bachman B,
tion. American Academy of Family Physicians, American
Roy-Burman P, Walker A, Mack TM. 1992. A revised estimate
Academy of Pediatrics, Advisory Committee on Immunization
of twin concordance in systemic lupus erythematosus. Arthritis
Practices. 2000. Summary of the joint statement on thimerosal
Rheum 35:311–318.
in vaccines. Public Health Service. Morb Mortal Wkly Rep
49:622– 631. De Flora S, Bennicelli C, Bagnasco M. 1994. Genotoxicity of
mercury compounds. A review. Mutat Res 317(1):57–79.
Cernichiari E, Brewer R, Myers GJ, Marsh DO, Lepham LW, Cox
C, Shamlaye CF, Berlin M, Davidson PW, Clarkson TW. 1995. DeGiorgio LA, Konstantinov KN, Lee SC, Hardin JA, Volpe BT,
Monitoring methylmercury during pregnancy: maternal hair Diamond B. 2001. A subset of lupus anti-DNA antibodies
predicts fetal brain exposure. Neurotoxicol 16:709 –713. cross-reacts with the NR2 glutamate receptor in systemic lupus
erythematosus. Nat Med 7(11):1175–1176.
[CHA] Child Health Alert. 2001. The Problem of mercury-con-
Dewailly E. 1997. Evaluation of prenatal exposure to organochlo-
taminated fish. Child Health Alert 19:3–5.
rine and heavy metals in Nunavik newborns 1993–1996. In:
Chan HM. 1998. Metal accumulation and detoxification in hu- Jense J, editor. Environmental Studies No. 74: synopsis of
mans. In: Langston WJ, Bebianno MJ, editors. Metal metabo- research conducted under the 1995–1997 Northern Contami-
lism in aquatic environments. London: Chapman and Hall. p nants Program. Ottawa, Ontario, Canada: Indian and Northern
415– 438. Affairs Canada. p 293–303.
Chang SW, Gilbert M, Sprecher J. 1978. Modification of methyl- Dianzani UM, Bragardo M, DiFranco D, Alliaudi C, Scagni P,
mercury neurotoxicity by vitamin E. Environ Res 17:356 –366. Buonfiglio D, Redoglia V, Bonissoni S, Correra A, Dianzani I,
Chang YC, Yeh C, Wang JD. 1995. Subclinical neurotoxicity of Ramenghi U. 1997. Deficiency of the Fas apoptosis pathway
mercury vapor revealed by a multimodality evoked potential without Fas gene mutations in pediatric patients with autoim-
study of chloralkali workers. Amer J Ind Med 27(2):271–279. munity/lymphoproliferation. Blood 891:2871–2880.
Chapman L, Chan HM. 2000. The influence of nutrition on methyl D’Itri RM. 1972. The Environmental mercury problem. Boca
mercury intoxication. Environ Health Perspect 108(1):29 –56. Raton, FL. CRC Press.
Choi BH, Lapham LW, Amin-Zaki L, Saleem T. 1978. Abnormal Drake CG, Rozzo SJ, Hirschfeld HF, Smarnworawong NP, Palmer
neuronal migration, deranged cerebral cortical organization and E, Kotzin BL. 1995. Analysis of the New Zealand Black con-
diffuse white matter astrocytosis of human fetal brain. A major tribution to lupuslike renal disease: multiple genes that operate
effect of methylmercury poisoning in utero. J Neuropathol Exp in a threshold manner. J Immunol 154:2441–2447.
Neurol 37:719 –733. Dubey C, Bellon B, Hirsch F, Kuhn J, Vial MC, Goldman M,
Chowdhury BA, Chandra RK. 1987. Biological and health impli- Druet P. 1991. Increased expression of class II major histocom-
cation of toxic heavy metal and essential trace element interac- patibility complex molecules on B cells in rats susceptible or
tions. Prog Food Nutr Sci 11:55–113. resistant to HgCl2-induced autoimmunity. Clin Exp Immunol
86:118 –127.
Clarkson T. 1991. Methylmercury. Fundam Appl Toxicol 16:20 –
Duits AJ, Bootsma H, Derksen RHWM, Spronk PE, Kater L,
31.
Kallenberg CGM, Capel PJA, Westerdaal NAC, Spierenburg
Clarkson TW. 1972. The pharmacology of mercury compounds. GTH, Gmelig-Meyling FHJ. 1995. Skewed distribution of IgG
Ann Rev Pharmacol 12:375– 406. Fc receptor Iia (CD32) polymorphism is associated with renal
Clarkson TW. 1997. The toxicology of mercury. Crit Rev Clin Lab disease in systemic lupus erythematosus patients. Arthritis
Sci 34:369 – 403. Rheum 39:1832–1836.
ENVIRONMENTAL EXPOSURE TO MERCURY 171

El-Begearmi MM, Ganther HE, Sunde ML. 1976. Vitamin E emy of Pediatrics, Committee on Environmental Health. Pedi-
decreases methylmercury toxicity. Poult Sci 55:2033–2042. atrics 108(1):197–205.
Eley BM. 1997. The future of dental amalgam: a review of the Goyer RA. 1996. Toxic effects of metals. In: Klaassen CD, editor.
literature. Part 5: Mercury in urine, blood and body organs from Casseret and Doull’s Toxicology. The Basic science of poisons.
amalgam fillings. Br Dent J 182:413– 417. 5th ed. New York: McGraw-Hill. p 691–736.
Elferink JG. 1999. Thimerosal: a versatile sulfhydryl reagent, Guery J, Druet E, Glotz D, Hirsch F, Mandet C, De Heer E, Druet
calcium mobilizer and cell function-modulating agent. Gen P. 1990. Specificity and cross-reactive idiotypes of anti-glomer-
Pharmacol 33(1):1– 6. ular basement membrane autoantibodies in HgCl2-induced au-
Endo T, Nakaya S, Kimura R, Murata T. 1984. Gastrointestinal toimmune glomerulonephritis. Eur J Immunol 20:93–107.
absorption of inorganic mercuric compounds in vivo and in situ. Hammond A. 1971. Mercury in the environment: natural and
Toxicol Appl Pharmacol 74:223–229. human factors. Science 171:788 –794.
Eneström S, Hultman P. 1995. Does amalgam affect the immune Hahn BH. 1994. Systemic Lupus Erythematosus: In: Isselbacher
system? A controversial issue. Int Arch Allergy Appl Immunol KJ, Braunwald E, Wilson JD, Martin J, Fauci AS, Kasper D,
106:180 –203. editors. Principles of internal medicine, Harrison 13th ed.
Ferber IA, Brocke S, Taylor-Edwards C, Ridgway W, Dinisco C, McGraw Hill, New York, p 1643–1648.
Steinman L, Dalton D, Fathmann CG. 1996. Mice with a dis- Hanley GA, Schiffenbauer J, Sobel ES. 1997. Class II haplotype
rupted IFN-␥ gene are susceptible to the induction of experi- differentially regulates immune response in HgCl2-treated mice.
mental autoimmune encephalomyelitis (EAE). J Immunol 156: Clin Immunol Immunopathol 84:328 –333.
5–13.
Hanley GA, Schiffenbauer J, Sobel ES. 2002. Resistance to
Fillastre J, Druet P, Mevy JP. 1998. Proteinuric nephropathies HgCl2-induced autoimmunity in haplotype-heterozygous mice
associated with drugs and substances of abuse. In: The Nephro- is an intrinsic property of B cells. J Immunol 161:1778 –1789.
pathic Syndrome. Cameron JS, Glassock eds. New York: John
Wiley & Sons. p 697–703. Hansen J, Danscher G. 1995. Quantitative and qualitative distri-
bution of mercury in organs from arctic sledgedogs: an atomic
Fisher GH, Rosenberg FJ, Straus SE, Dale JK, Middleton LA, Lin
absorption spectrophotometric and histochemical study of tissue
AY, Strober W, Lenardo MJ, Puck JM. 1995. Dominant inter-
samples from natural long-termed high dietary organic mercury-
fering Fas gene mutations impair apoptosis in a human autoim-
exposed dogs from Thule, Greenland. Pharmacol Toxicol 77:
mune lymphoproliferative syndrome. Cell 81:935–947.
189 –195.
Fitzgerald WF, Clarkson TW. 1991. Mercury and monomethyl
Harada M, Nakanishi J, Konuama S, Ohno K, Kimura I, Yamagu-
mercury: present and future concerns. Environ Health Perspect
chi H, Tsuruta K, Kizaki T, Ookawara T, Ohno H. 1998. The
96:159 –166.
present mercury contents of scalp hair and clinical symptoms in
Frustaci A, Magnavita N, Chimenti C, Caldarulo M, Sabbioni E, inhabitants of the Minamata. Environ Res 77:160 –164.
Pietra R, Cellini C, Possati FG, Maseri S. 1999. Marked eleva-
Harris S, Wilson J, Printz R. 1973. Embryotoxicity of methyl
tion of myocardial trace elements in idiopathic dilated cardio-
mercuric chloride in golden hamsters. Teratology 6:139 –148.
myopathy. J Am Coll Cardiology 33(6):1578 –1583.
Fujinka NS, Zasshi GF. 1987. Binding of Hg2⫹ to transmembrane Hirsch F, Couderc J, Sapin C, Fournie G, Druet P. 1982. Poly-
thiol groups was implicated in the decline of the sugar-Na⫹ clonal effect of HgCl2 in the rat, its possible role in an experi-
phlorizin-sensitive cotransport system, and this inhibited galac- mental autoimmune disease. Eur J Immunol 12:620 – 631.
tose absorption in rats [in Japanese]. Acta Obstetrica et Gynae- Hirsch F, Kuhn J, Ventura M, Vial M-C, Fournie G, Druet P. 1986.
cologica Japonica 39:2133–2136. Autoimmunity induced by HgCl2 in brown Norway rats. I.
Fukino H, Hirai M, Hsueh YM, Moriyasu S, Yamane Y. 1986. Production of monoclonal antibodies. J Immunol 36(9):3272–
Mechanism of protection by zinc against mercuric chloride 3276.
toxicity in rats: effects of zinc and mercury on glutathione Hogarth MB, Slingsby JH, Allen PJ, Thompson EM, Chandler P,
metabolism. J Toxicol Environ Health 19:75– 89. Davies KA, Simpson E, Morley BJ, Walport MJ. 1998. Multiple
Galal-Gorchev H. 1993. Dietary intake, levels in food and esti- lupus susceptibility loci map to chromosome 1 in BXSB mice.
mated intake of lead, cadmium, and mercury. Food Addit Con- J Immunol 161:2753–2764.
tam 10:115–128. Hojbjerg S, Nielsen JB, Anderson O. 1992. Effects of dietary
Gale TF. 1984. The amelioration of mercury-induced embryotox- lipids on whole-body retention and organ distribution of organic
icity effects by simultaneous treatment with zinc. Environ Res and inorganic mercury in mice. Food Chem Toxicol 30:703–
35:405– 412. 708.
Gilbert SG, Grant-Webster K. 1995. Neurobehavioral effects of Hojbjerg SG. 1996. The effect of nutritional factors on absorption,
developmental methylmercury exposure. Environ Health Per- retention and excretion of organic and inorganic mercury in
spect 103(6):135–142. mice and rats. Danish Med Bull 43:376 –384.
Gillespie KM, Qasim J, Tibbatts LM, Thiru S, Oliveira DB, Hu H. 2000. Exposure to metals. Occupational Env Med 27:983–
Mathieson PW. 1995. Interleukin-4 gene expression in mercury- 996.
induced autoimmunity. Scand J Immunol 41:268 –277. Hultgren B, Huang X, Dybdal N, Stewart TA. 1996. Genetic
Goldman LR, Shannon MW. 2001. Technical report: mercury in absence of gamma-interferon delays but does not prevent dia-
the environment: implication for pediatricians. American Acad- betes in NOD mice. Diabetes 45:812– 819.
172 TCHOUNWOU ET AL.

Hultman P, Eneström S. 1987. The induction of immune complex Kono DH, Burlingame RW, Owens DG, Kuramochi A, Balderas
deposits in mice by peroral and parenteral administration of RS, Balomenos D, Theophilus AN. 1994. Lupus susceptibility
mercuric chloride: strain-dependent susceptibility. Clin Exp Im- loci in New Zealand mice. Proc Natl Acad Sci USA 91:10168 –
munol 67:28 –35. 10172.
Hultman P, Bell LJ, Enestrom S, Pollard KM. 1992. Murine Kono H, Park MS, Szydlik A, Haraldsson K, Kuan JD, Pearson
susceptibility to mercury. I. Autoantibody profiles and systemic PL, Hultman P, Pollard KM. 2001. Resistance to xenobiotic-
immune deposits in inbred, congenic and intra-H-2 recombinant induced autoimmunity maps to chromosome 1. J Immunol 167:
strains. Clin Immunol Immunopathol 65:98 –108. 2396 –2403.
Hultman P, Bell LJ, Eneström S, Pollard KM. 1993. Murine Koos BJ, Longo LD. 1976. Mercury toxicity in the pregnant
susceptibility to mercury. II. Autoantibody profiles and renal woman, fetus, and newborn infant: a review. Am J Obstet
immune deposits in hybrid, backcross, and H-2d congenic mice. Gynecol 126:390 – 409.
Clin Immunol Immunopathol 68:9 –17. Kulig K. 1998. A tragic reminder about organic mercury. N Engl
Hultman P, Eneström S, Turley SJ, Pollard KM. 1994. Selective J Med 338:1692–1694.
induction of anti-fibrillarin autoantibodies by silver nitrate in Kuznetsov DA. 1987. Paradoxical effect of methyl mercury on
mice. Clin Exp Immunol 96:285–293. mitochondrial protein synthesis in mouse brain tissue. Neuro-
Hultman P, Johansson U, Dagnaes-Hansen F. 1995a. Murine mer- chem Res 12:751–753.
cury-induced autoimmunity: the role of T-helper cells. J Auto-
Langford N, Ferner R. 1999. Toxicity of mercury. J Hum Hyper-
immun 8:809 – 817.
tens 13:651– 656.
Hultman P, Ganowiak K, Turley SJ, Pollard KM. 1995b. Genetic
Langworth S, Elinder C-G, Göthe C-J, Vesterberg O. 1991. Bio-
susceptibility to silver-induced anti-fibrillarin autoantibodies in
logical monitoring of environmental and occupational exposure
mice. Clin Immunol Immunopathol 77:291–303.
to mercury. Int Arch Occup Environ Health 46:102–109.
Hultman P, Lindh U, Horsted-Bindsev P. 1998. Activation of the
LeBel CP, Ali SF, Bondy SC. 1992. Deferoxamine inhibits meth-
immune system and systemic immune-complex deposits in
yl-mercury induced increases in reactive oxygen species forma-
brown Norway rats with dental amalgam restorations. J Dent
tion in rat brain. Toxicol Appl Pharmacol 112:161–165.
Res 77:1415–1425.
Le Deist F, Emile J-F, Rieux-Laucat F, Benkerrou MI, Roberts I,
Inouye M, Hoshino K, Murakami U. 1972. Effect of methylmer-
Brousse N, Fischer A. 1996. Clinical, immunological, and
curic chloride on embryonic and fetal development in rats and
pathological consequences of Fas-deficient conditions. Lancet
mice. Ann Rep Res Inst Environ Med Nagoya Univ 19:69 –78.
348:719 –726.
Jelili MA, Abbasi AH. 1981. Poisoning by ethyl mercury toluene
sulfonanilide. Br J Ind Med 16:303–308. Leonard MD, Chater BV, Duffield DP, O’Sullivan JJ, Lockwood
CM. 1983a. Circulating immune complexes in individuals oc-
Jiang YG, Möller G. 1995. In vitro effects of HgCl2 on murine
cupationally exposed to mercury vapor. In: Brown SS, Savory J,
lymphocytes. I. Preferable activation of CD4⫹ T cells Ia re-
editors. Chemical toxicology and clinical chemistry of metals
sponder strain. J Immunol 154:3138 –3143.
135: New York: Academic Press.
Jones LS, Rizzo LV, Agarwal RK, Tarrant TK, Chan CC, Wiggert
Leonard A, Jacquet P, Lauwerys RR. 1983b. Mutagenicity and
B, Caspi RR. 1997. IFN-␥-deficient mice develop experimental
teratogenicity of mercury compounds. Mutat Res 114(1):1–18.
autoimmune uveitis in the context of a deviant effector response.
J Immunol 158:5997– 6008. Levander OA, Cheng L. 1980. Micronutrient interactions, vita-
mins, minerals, and hazardous elements. Ann NY Acad Sci
Kass S, Tyc K, Steitz JA, Sollner Webb B. 1990. The U3 small
355:1–372.
nucleolar ribonucleoprotein functions in the first step of preri-
bosomal RNA processing. Cell 60:897–903. Lin JL, Lim PS. 1993. Massive oral ingestion of elemental mer-
cury. J Toxicol Clin Toxicol 31:487– 492.
Kasturi KN, Hatakeyama A, Spiera H, Bona CA. 1995. Antifibril-
larin autoantibodies present in systemic sclerosis and other Lodenius M, Malm O. 1998. Mercury in the Amazon. Rev Environ
connective tissue diseases interact with similar epitopes. J Exp Contam Toxicol 157:25–52.
Med 181:1027–1034. Loppi S, Bonini I. 2000. Lichens and mosses as biomonitors of
Kerper LE, Belletori N, Clarkson TW. 1992. Methylmercury trans- trace elements in areas with thermal springs and fumarole ac-
port across the blood– brain barrier by an amino acid carrier. tivity (Mt. Amiata, central Italy). Chemosphere 41:1333–1336.
Am J Physiol 207:R761–R765. Lubben B, Rottman N, Kubicki Muranyi M, Gleichmann E, Lu-
King LJ, Soares JH. 1981. The effect of vitamin E and dietary hrmann R. 1994. The specificity of disease-associated anti-
linoleic acid on mercury toxicity. Nutr Rep Int 24:39 – 45. fibrillarin autoantibodies compared with that of HgCl2-induced
Klassen CD. 1990. Heavy metals and heavy-metal antagonists. In: autoantibodies. Mol Biol Rep 20:63–71.
Gilman AC, Rall TW, Niew AS, Taylor P, editors: The phar- Lugea A, Barber A, Ponz F. 1994. Inhibition of D-galactose and
macological basis of therapeutics. 8th ed. New York: Pergamon L-phenylalanine transport by HgCl2 in rat intestine in vitro.
Press, p 1598 –1602. Revista Espaniola Fisiolog 50:167–173.
Kono DH, Balomenos D, Pearson DL, Park MS, Gildebrandt B, Magos L. 1991. Overview on the protection given by selenium
Hultman P, Pollard KM. 1998. The prototypic TH2 autoimmu- against mercurials. In: Suzuki T, Imura N, Clarkson TW, edi-
nity induced by mercury is dependent on IFN-␥ and not TH1/ tors. Advances in mercury toxicology. New York: Plenum
TH2 imbalance. J Immunol 161:234 –240. Press.
ENVIRONMENTAL EXPOSURE TO MERCURY 173

Magos L, Clarkson TW, Hudson AR. 1984. Differences in the mercury-exposed mice. Effect of seleno-L-methionine. Toxicol
effects of selenite and biological selenium on the chemical form Lett 76:165–171.
and distribution of mercury after the simultaneous administra- Nierenberg DW, Nordgren RE, Chang MB, Siegler RW, Blayney
tion of HgCl2 and selenium to rats. J Pharmacol Exp Ther MB, Hochberg F, Toribara TY, Cernchiari E, Clarkson T. 1998.
228:478 – 483. Delayed cerebellar disease and death after accidental exposure
Manoury-Schwartz B, Chiocchia G, Bessis N, Abehsira-Amar O, to dimethylmercury. N Engl J Med 338:1672–1676.
Batteux F, Muller S, Huang S, Boissier MC, Fournier C. 1997. Norseth T. 1970. Biliary excretion and intestinal reabsorption of
High susceptibility to collagen-induced arthritis in mice lacking mercury in the rat after injection of methyl mercuric chloride.
IFN-␥ receptors. J Immunol 158:5501–5506. Acta Pharmacol Toxicol 33:280 –288.
Marsh DO, Clarkson TW, Myers GJ, Davidson PW, Cox C, Norseth T, Clarkson TW. 1970. Studies on the biotransformation
Cermichiari E, Tanner MA, Lednar W, Shamlaye C, Choisy O. of 203Hg-labeled methyl mercury chloride in rats. Arch Environ
1995. The Seychelles study of fetal methylmercury exposure Health 21:717–727.
and child development: introduction. Neurotoxicol 16:583–596. Nriagu JO, Pacyna J. 1988. Quantitative assessment of worldwide
Mathieson DS, Clarkson TW, Gelfand EW. 1980. Mercury toxic- contamination of air, water and soils by trace metals. Nature
ity (acrodyna) induced by long-term injection of gamma glob- 333:134 –139.
ulin. J Pediatr 97:153–155. Nygard NR, McCarthy DM, Schiffenbauer J, Schwartz BD. 1993.
Mathieson PW, Thiru SD, Oliveira DB. 1992. Mercuric chloride- Mixed haplotypes and autoimmunity. Immunol Today 14:53–
treated brown Norway rats develop widespread tissue injury 61.
including necrotizing vasculitis. Lab Invest 67:121–132. Ochel M, Vohr HW, Pfeiffer C, Gleichmann E. 1991. IL-4 is
Mirtcheva J, Pfeiffer C, De Bruijn JA, Jacquesmart F, Gleichmann required for the IgE and IgG1 increase and IgG1 autoantibody
E. 1989. Immunological alterations inducible by mercury com- formation in mice treated with mercuric chloride. J Immunol
pounds. III. H-2A acts as an immune response and H-2e as an 146:3006 –3015.
immune “suppression” locus for HgCl2-induced antinuclear au- Pelletier L, Pasquier R, Hirsch F, Sapin C, Druet P. 1986. Auto-
toantibodies. Eur J Immunol 19:2257–2268. reactive T cells in mercury-induced autoimmune disease: in
Mokrzan EM, Kerper LE, Ballatori N, Clarkson TW. 1995. Meth- vitro demonstration. J Immunol 137:2548 –2557.
ylmercury–thiol uptake into cultured brain capillary endothelial Philbert MA, Billingsley ML, Reuhl KR. 2000. Mechanisms of
cells on amino acid system L. J Pharmacol Exp Ther 272:1277– injury in the central nervous system. Toxicol Pathol 26(1):43–
1284. 53.
Morel L, Rudofsky UH, Longmate JA, Schiffenbauer J, Wakeland Pier SM. 1975. The role of heavy metals in human health. Texas
EK. 1994. Polygenic control of susceptibility to murine sys- Rep Biol Med 33(1):85–106.
temic lupus erythematosus. Immunity 1:219 –229. Pietsch P, Vohr H-W, Degitz K, Gleichmann E. 1989. Immuno-
Moser KL, Gray-McGuire C, Kelly J, Asundi N, Yu H, Bruner logical alterations inducible by mercury compounds. II. HgCl2
GR, Mange M, Hogue R, Neas BR, Harley JB. 1999. Confir- and gold sodium thiomalate enhance serum IgE and IgG con-
mation of genetic linkage between human systemic lupus ery- centrations in susceptible mouse strains. Int Arch Allergy Appl
thematosus and chromosome 1q41. Arthritis Rheum 42:1902– Immunol 90:47–53.
1915. Pollard KM, Hultman P. 1997. Effects of mercury on the immune
Moser KL, Neas BR, Salmon JE, Yu H, Gray-McGuire C, Asundi system. In: Sigel H, Sigel A, editors. Metal ions in biological
N, Bruner GR, Fox J, Kelly J, Henshall S, Basino D, Dietz M, systems. Mercury and its effects on environment and biology.
Hogue R, Koelsch G, Nightingale L, Shaver T, Abdou NI, New York: Marcel Dekker: 421– 440.
Albert DA, Carson C, Petri M, Treadwell EL, James JA, Harley Pollard KM, Lee DK, Casiano CA, Blüthner M, Johnson MM, Tan
JB. 1998. Genome scan of human systemic lupus erythemato- EM. 1997. The autoimmunity-inducing xenobiotic mercury in-
sus: evidence for linkage on chromosome 1q in African-Amer- teracts with the autoantigen fibrillarin and modifies its molecular
ican pedigrees. Proc Natl Acad Sci USA 95:14869 –14874. and antigenic properties. J Immunol 158:3521–3528.
Mosmann TR, Cherwinski H, Bond MW, Giedlin A, Coffman RL. Pollard KM, Pearson LD, Blüthner M, Tan EM. 2002. Proteolytic
1986. Two types of murine helper T cell clone. I. Definition cleavage of a self-antigen following xenobiotic-induced cell
according to profiles of lymphokine activities and secreted pro- death produces a fragment with novel immunogenic properties.
teins. J Immunol 136:2348 –2356. J Immunol 165:2263–2270.
Myers GJ, Davidson PW, Cox A, Shamlaye CF, Tanner MA, Pollard KM, Pearson DL, Hultman P, Deane TN, Lindh U, Kono
Choisy O, Sloane-Reeves J, Marsh D, Cernichiari E, Choi A. DH. 2001. Xenobiotic acceleration of idiopathic systemic auto-
1995. Neurodevelopmental outcomes of Seychellois children immunity in lupus prone BXSB mice. Environ Health Perspect
sixty-six months after in utero exposure to methylmercury from 109:27–35.
a maternal fish diet: pilot study. Neurotoxicol 16:639 – 652. Pollard KM, Pearson DL, Hultman P, Hildebrandt B, Kono DH.
Mykkanen HM, Metsanitty L. 1987. Selenium–mercury interac- 1999. Lupus-prone mice as models to study xenobiotic-induced
tion during intestinal absorption of 75Se compounds in chicks. J acceleration of systemic autoimmunity. Environ Health Perspect
Nutr 117:1453–1458. 107:729 –735.
Nielsen JB, Andersen O. 1995. A comparison of the lactational and Prati M, Gornati R, Boracchi P, Biganzoli E, Fortaner S, Pietra R,
transplacental deposition of mercury in offspring from methyl- Sabbioni E, Bernardini G. 2002. A comparative study of the
174 TCHOUNWOU ET AL.

toxicity of mercury dichloride and methylmercury, assayed by Sallustro F, Mackay CR, Lanzavecchia A. 1997. Selective expres-
the frog embryo teratogenesis assay-xenopus (FETAX). Altern sion of the eotaxin receptor CCR3 by human helper 2 cells.
Lab Anim 30(1):23–32. Science 277:2005–2014.
Puck JM, Sneller MC. 1997. ALPS: An autoimmune human lym- Salmon JE, Millard S, Schachter LA, Arnett FC, Ginzler EM,
phoproliferative syndrome associated with abnormal lympho- Gourley MF, Ramsey-Goldman R, Peterson MG, Kimberly RP.
cyte apoptosis. Semin Immunol 9:77– 81. 1996. Fc␥RIIA alleles are heritable risk factors for lupus ne-
Rana SV, Sharma R. 1982. Co-enzyme effects of inorganic mer- phritis in African-Americans. J Clin Invest 97:1348 –1354.
cury in the liver of a freshwater fish Channa punctatus. J Appl Sapin C, Hirsch F, Delaporte JP, Bazin H, Druet P. 1984. Poly-
Toxicol 2:275–277. clonal IgE increase after HgCl2 injections in BN and LEW rats:
Rice G, Mahaffery K, Lyon B. 1999. Predicting exposure: U.S. a genetic analysis. Immunogenetics 20:227–234.
EPA. Mercury Report to Congress. In: Mercury in Eastern Sapin C, Mandet C, Druet E, Gunther G, Druet P. 1981. Immune
Canada and Northeast states. Proceedings of the Conference, complex type disease induced by HgCl2; genetic control of
21–23 September 1998, Fredericton, New Bruswick, Ontario, susceptibility. Transplant Proc 13:1404 –1413.
Canada. Schuhmann D, Kubicka-Muranyi M, Mirtschewa J, Gunther J,
Richardson DHS. 1992. Pollution monitoring with lichens. Slough, Kind P, Gleichmann E. 1990. Adverse immune reactions to
VA: Richmond Publishing. gold. I. Chronic treatment with an Au(I) drug sensitizes mouse
Roberts H, Leonard D, Osborne J. 2001. Potential health and T cells not to Au(I), but to Au(III) and induces autoantibody
environmental issues of mercury-contaminated amalgamators. formation. J Immunol 145:2123–2132.
J Am Dent Assoc 132:58 – 64. Schwarz S, Hussedt I, Bertram HP, et al. 1996. Amyotropic lateral
Roether S, Rabbani H, Mellstedt H, Abedi-Valugerdi M. 2002. sclerosis after accidental injection of mercury. J Neurol Neuro-
Spontaneous downregulation of antibody/autoantibody synthe- surg Psychiatry 60:698 –702.
sis in susceptible mice upon chronic exposure to mercuric Sercarz EE, Lehmann PV, Ametani A, Benichou G, Miller A,
chloride is not owing to a general immunosuppression. Scand Moudgil K. 1993. Dominance and crypticity of T cell antigenic
J Immunol 55:493–502. determinants. Annu Rev Immunol 11:729 –737.
Röger J, Zillikens D, Burg G, Gleichmann E. 1992. Systemic Shi X, Xie C, Kreska D, Richardson JA, Mohan C. 2002. Genetic
autoimmunity in a patient with longstanding exposure of mer- dissection of SLE, SLE1 and FAS impact alternate pathways
cury. Eur J Dermatol 2:168 –177. leading to lymphoproliferative autoimmunity. J Exp Med 196:
Roman-Franco AA, Turiello M, Albini B, Ossi E, Milgrom F, 281–292.
Andres GA. 1978. Anti-basement membrane antibodies and Solomons NW, Viteri FE. 1982. Biological interaction of ascorbic
antigen–antibody complexes in rabbits injected with mercuric acid and mineral nutrients iron, selenium, copper, nickel, man-
chloride. Clin Immunol Immunopathol 9:464 – 472. ganese, zinc, cobalt, cadmium, mercury, vitamin C. Adv Chem
Roths JB, Murphy ED, Eicher EM. 1984. A new mutation, Gld, Ser 200:551–569.
that produces lymphoproliferation and autoimmunity in C3H/ Steinnes E, Krog H. 1977. Mercury, arsenic and selenium fall-out
HeJ mice. J Exp Med 159:1–11. from an industrial complex studied by means of lichen trans-
Rouleau C, Gobeil C, Tjälve H. 2000. Accumulation of silver from plants. Oikos 28:160 –164.
the diet in two marine benthic predators: the snow crab (Chio- Stem AH. 1993. Re-evaluation of the reference dose for methyl-
noectetes opilio) and American plaice (Hippoglossoides plates- mercury and assessment of current exposure levels. Risk Anal
soides). Environ Toxicol Chem 19:631– 637. 13:355–364.
Rowland I, Davies M, Evans J. 1980. Tissue content of mercury in Stiller-Winkler R, Radaszkiewicz T, Gleichmann E. 1988. Immu-
rats given methyl mercury chloride orally: influence of intestinal nopathological signs in mice treated with mercury compounds.
flora. Arch Environ Health 25:155–160. I. Identification by the popliteal lymph node assay of responder
Rowland IR, Mallett AK, Flynn J, Hargreaves RJ. 1986. The effect and nonresponder strains. Int J Immunopharm 10:475– 482.
of various dietary fibers on tissue concentration and chemical Suda I, Hirayama K. 1992. Degradation of methyl and ethyl
form of mercury after methylmercury exposure in mice. Arch mercury not inorganic mercury by hydroxyl radical produced
Toxicol 59:94 –98. from rat liver microsomes. Arch Toxicol 66(1):34 – 41.
Rowland IR, Robinson RD, Doherty RA. 1984. Effects of diet on Suda I, Takahashi H. 1990. Effect of reticuloendothelial system
mercury metabolism and excretion in mice given methylmer- blockade on the biotransformation of methylmercury in the rat.
cury: role of gut flora. Arch Environ Health 39:401– 408. Bull Environ Contam Toxicol 44:609 – 615.
Rumbeiha WK, Gentry PA, Bhatnagar MK. 1992. The effects of Sutton D, Tchounwou PB, Nanuli N, Shen E. 2002. Mercury
administering methylmercury in combination with ethanol in the induces cytotoxicity and transcriptionally activates stress genes
rat. Vet Human Toxicol 34:21–25. in human liver carcinoma (HepG2) cells. Int J Mol Sci 3:962–
Sager PR, Aschner M, Roder PM. 1984. Persistent differential 981.
alterations in developing cerebellar cortex of male and female Suzuki T, Tekemoto T-I, Kashiwazaki H, Miyama T. 1973. Met-
mice after methylmercury exposure. Dev Brain Res 12:1–11. abolic fate of ethylmercury salts in man and animal. In: Miller
Sakamoto M, Ikegami N, Nakano A. 1996. Protective effects of MW, Clarkson TW, editors. Mercury, Mercurials and Mercap-
Ca2⫹ channel blockers against methyl mercury toxicity. Phar- tans. Springfield, IL: Charles C Thomas Publishers. p 209 –232.
macol Toxicol 78:193–199. Sweet LL. 2001. Toxicology and immunology of mercury—a
ENVIRONMENTAL EXPOSURE TO MERCURY 175

comparative review in fish and humans. J Toxicol Environ Vermeire K, Heremans H, Vandeputte M, Huang S, Billiau A,
Health 4(2):161–203. Matthys P. 1997. Accelerated collagen-induced arthritis in
Szabo SJ, Dighe AS, Gubler U, Murphy KM. 1997. Regulation of IFN-␥ receptor-deficient mice. J Immunol 158:5507–5513.
the interleukin (IL)-12Rb2 subunit expression in developing T Vijayalakshmi K, Apu C, Sood PP. 1992. Differential effects of
helper1 (Th1) and Th2 cells. J Exp Med 185:817– 828. methylmercury, thiols and vitamins on galactosidases of ner-
Takahashi T, Tanaka M, Brannan CI, Jenkins NA, Copeland NG, vous and non-nervous tissues. Bull Environ Contam Toxicol
Suda T, Nagata S. 1994. Generalized lymphoproliferative dis- 49:71–77.
ease in mice, caused by a point mutation in the Fas ligand. Cell Watanabe-Fukunaga R, Brannan CI, Copeland NG, Jenkins NA,
76:969 –979. Nagata S. 1992. Lymphoproliferation disorder in mice ex-
Takeuchi K, Turley SJ, Tan EM, Pollard LM. 1995. Analysis of plained by defects in Fas antigen that mediates apoptosis. Nature
the autoantibody response to fibrillarin in human disease and 356:314 –326.
murine models of autoimmunity. J Immunol 154:961–969. Welsh SO. 1977. Contrasting effects of vitamins A and E on
Tan M, Perkin JE. 2000. Route of decomposition of thiomerosal mercury poisoning. Fed Proc 36:1146 –1156.
(thimerosal). Int J Pharm 208(1–2):23–34. Welsh SO, Soares JH. 1975. Effects of selenium and vitamin E on
Tchounwou PB, Ishaque AB, Sutton D, Ninashvili N, Shen E. methyl mercury toxicity in the Japanese quail. Fed Proc 34:913–
2002. Biomarkers of sensitivity and effect associated with cad- 919.
mium and mercury toxicity in human liver carcinoma (HepG2) Whitekus MJ, Santini RP, Rosenspire AJ, McCabe MJ Jr. 1999.
cells. In: Khassanova I, Collery P, Maynard I, Khassanova Z, Protection Against CD95-mediated apoptosis by inorganic mer-
Etienne JC, editors. Metal ions in biology and medicine: Paris: cury in Jurkat T cells. J Immunol 162:7162–7170.
John Libbey Eurotext. Vol. 7, p 132–137. [WHO] World Health Organization. 1990. Methylmercury. Inter-
Theofilopoulos AN. 1995. The basis of autoimmunity: Part 1. national Program on Chemical Safety. Geneva, Switzerland:
Immunol Today 16:90 –98. World Health Organization. Environmental Health Criteria 101:
Theofilopoulos AN, Kono DH. 1999. Murine lupus models: gene- 1–144.
specific and genome-wide studies. In: Lahita RG, editor. Sys- Woshner VM, O’Hara TM, Eurell JA, Wallig MA, Bratton GR,
temic lupus erythematosus. 3rd ed. New York: Churchill Liv- Suydam RS, Beasley VR. 2002. Distribution of inorganic mer-
ingstone. p 149 –181. cury in liver and kidney of beluga and bowhead whales through
Tominack R, Weber J, Blume C, Madhok M, Murphy T, Thomp- autometallographic development of light microscope tissue sec-
son M. 2002. Elemental mercury as an attractive nuisance: tions. Toxicologic Pathology 30(2):209 –215.
multiple exposures from a pilfered school supply with severe Woshner VM, O’Hara TM, Bratton GR, Suydam RS, Beasley VR.
consequences. Pediatr Emerg 18(2):97–100. 2001. Concentrations and interactions of selected essential and
Tsao BP, Cantor RM, Kalunian KC, Chen C-J, Badsha H, Singh R, non-essential elements in bowhead and beluga whales of
Wallace DJ, Kitridou RC, Chen S-L, Shen N, Song YW, Isen- Alaska. J Wild Dis 37:693–710.
berg DA, Yu C-L, Hahn BH, Rotter JI. 1997. Evidence for Yannai S, Sachs KM. 1993. Absorption and accumulation of
linkage of a candidate chromosome 1 region to human systemic cadmium, lead and mercury from foods by rats. Food Chem
lupus erythematosus. J Clin Invest 99:725–731. Toxicol 31:351–355.
Urbach J, Boadi W, Brandes JM, Kerner H, Yannai S. 1992. In Yasutake A, Hirayama K, Inouye M. 1990. Interaction of methyl-
vitro effect of mercury on enzyme activities and its accumula- mercury compounds with albumin. Arch Toxicol 64:639 – 643.
tion in first-trimester human placenta. Environ Res 57:96 –106. Zhang J. 1984. Clinical observations in ethyl mercury chloride
[U.S. EPA] Environmental Protection Agency. 1997. Mercury. poisoning. Am J Ind Med 5:251–255.
Report to Congress Office of Air Quality and Standards. Wash- Zheng W, Flavell R. 1997. The transcription factor GATA-3 is
ington DC: U.S. Environmental Protection Agency. necessary and sufficient for Th2 cytokine gene expression in
Van Vliet E, Uhrberg M, Stein C, Gleichmann E. 1993. MHC CD4 T cells. Cell 89:587–594.
control of IL-4-dependent enhancement of B cell Ia expression Zorn NE, Smith JT. 1990. A relationship between vitamin B12,
and Ig class switching in mice treated with mercuric chloride. folic acid, ascorbic acid, and mercury uptake and methylation.
Int Arch Allergy Immunol 101:392– 402. Life Sci 47(2):167–173.

S-ar putea să vă placă și