Sei sulla pagina 1di 10

Review Article

Indian J Med Res 128, October 2008, pp 373-382

Methylmercury neurotoxicity & antioxidant defenses

José Luiz M. do Nascimento1, Karen Renata M. Oliveira1, Maria Elena Crespo-Lopez2, Barbarella M.
Macchi1, Luís Antônio L. Maués 1, Maria da Conceição N. Pinheiro 3, Luiz Carlos L. Silveira3,4 &
Anderson Manoel Herculano 1,5

1
Lab. Neuroquímica Molecular e Celular, Dep. Fisiologia, Instituto de Ciências Biológicas
2
Lab. Farmacologia Molecular, Instituto de Ciências Biológicas, 3Núcleo de Medicina Tropical
4
Lab. Neurofisiologia, Instituto de Ciências Biológicas, & 5Lab. Neuroendocrinologia Instituto de
Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil

Received April 21, 2008

Neurotoxicity induced by methylmercury (MeHg) increases the formation of reactive radicals and
accelerates free radical reactions. This review summarizes recent findings in the MeHg- induced
formation of free radicals and the role of oxidative stress in its neurotoxicity. Oxidative stress on
CNS can produce damage by several interacting mechanisms, including mitochondrial damage with
increase in intracellular free Ca2+, activation and inhibition of enzymes, release of excitatory amino
acids, metallothioneins expression, and microtubule disassembly. The nature of antioxidants is
discussed and it is suggested that antioxidant enzymes and others antioxidants molecules may protect
the central nervous system against neurotoxicity caused by MeHg.

Key words Anti-oxidant defenses - metallothioneins - methylmercury - microtubules network - neurotransmitters - oxidative stress

Methylmercury toxicity exposed individuals exhibit severe forms of


neurological disease which include a collection of
Mercury is a heavy metal contaminant with
cognitive, sensory, and motor disturbance5,6.
potential for global mobilization following its give off
from anthropogenic activities or natural processes1. The studies on MeHg toxicity have tried to evaluate
In nature, elemental mercury (Hg 0 ) can be its impact on several ecosystems around the World,
biotransformed and converted to methylmercury including places in Japan, Irak, Canada, Africa,
(MeHg), which is the most toxic form of mercury in including Brazilian Amazon, and India2,7,8, as well as to
the environment2-4. The studies about MeHg toxicity understand its toxicological effect on biological
became ubiquitous and diversified since the outbreak systems. MeHg was firstly recognized as a potent
of environmental catastrophes such as those in neurotoxicant for the adult nervous system in studies
Minamata (1950s) and Niigata (1960s). In such performed on exposed workers of a chemical factory in
episodes, as a consequence of MeHg exposure, the England9,10. Later, its importance as a neurotoxicant for
373
374 INDIAN J MED RES, OCTOBER 2008

the nervous system during development was recognized the presence of such transporters is crucial for toxicity
in the Minamata’s outbreak5,6. Since then, several studies to occur at least through this mechanism, transporter
of exposed human populations as well as experiments inhibition is expected to be beneficial to prevent
with laboratory animals demonstrated that exposure to disorders caused by MeHg toxicity.
toxic levels of MeHg during pre- and post-natal life
Cellular mechanism to MeHg intoxication
causes neurological abnormalities, cognitive
impairment, and behavioural disturbance11,12. MeHg Calcium homeostasis: Calcium ion (Ca2+) plays a critical
vulnerability of the developing brain reflects the ability role in CNS cell death. Ca 2+ increase beyond
of lipophylic MeHg to cross the placenta and to physiological levels activates catabolic enzymes such
concentrate in the central nervous system (CNS) once as phospholipases, proteases, and endonucleases, causes
the blood-brain barrier is not fully developed in the mitochondrial dysfunction, and disturbes cytoskeletal
prenatal period13,14. organization. Several lines of evidence indicate that at
low concentrations MeHg disrupts Ca2+ homeostasis,
The developing CNS is more affected by MeHg increasing its intracellular level in a number of
exposure and exhibits different patterns of changes than experimental situations, including primary culture of
the mature CNS. In the adult brain, MeHg poisoning cerebellar granule cells21. This effect has all the potential
damages the so-called primary areas of the cerebral to disrupt the synaptic function and impair the neural
cortex, affecting the visual, auditory, somatic sensory, development22.
and motor cortex, as well as the hippocampus and the
granule layer of the cerebellum, causing a remarkable Ca 2+ metabolism is altered through specific
loss of neurones in these brain regions. On the other pathways which affect Ca 2+ regulation by some
hand, in the developing brain there is a widespread organelles such as mitochondria and smooth
neuronal loss throughout the CNS, what has been endoplasmic reticulum (SER)23. Ca2+ channel blockers
interpreted as due to the high MeHg sensitivity of the significantly delay MeHg-induced increase of Ca2+
immature CNS 11. The neural disease due to MeHg levels in vitro24. In agreement with these data, blockers
neurotoxicity includes such symptoms as visual field of voltage-dependent Ca2+ channels have been shown
constriction, somatic sensory disturbance, hearing to prevent the appearance of neurological disorders in
disturbance, cerebellar ataxia, dysarthria, and mnemic rats administered with MeHg25. These observations give
deficits15-17. support to the hypothesis that changes in Ca 2+
homeostasis represent an important cellular event in the
Membrane interactions and transporter-mediated MeHg-induced CNS toxicity.
MeHg toxicity
Mitochondrial damage induced by MeHg: Mitochondria
MeHg quickly diffuses across membranes without are the main intracellular site for reactive oxygen
significant partitioning in lipid bilayers. Thus, it has production and one of the most susceptible targets for
been proposed that MeHg toxicity is mediated by MeHg radical species to exert their actions. Importance of
membrane leakage 17 . However, it has also been mitochondria for MeHg toxicity was recognized from
suggested that the potential of MeHg to increase studies performed both in vivo and in vitro. In vivo
oxidative events leading to cell damage is controlled exposure to MeHg causes its accumulation inside
by MeHg binding to membrane transporters. MeHg mitochondria followed by a series of biochemical
absorption, distribution, and excretion are commonly changes in these organelles26. These effects are similar
mediated by plasma membrane protein transporters18. to those observed in studies of mitochondria respiratory
In addition, it has been possible to investigate at chain inhibition27.
molecular level the mechanisms of MeHg transport
Under physiological conditions, the electron
through membrane transporters with broad substrate
transport system of mitochondria consumes about 1-5
selectivity. These transporters are known as
per cent of the oxygen that is converted to reactive
“multispecific”, taking advantage of its nature to exert
oxygen species (ROS) such as O2 and H2O227. Animals
their toxic effects 18 . The main route for MeHg
exposed to MeHg display neurological symptoms after
transmembrane transport seems to be the amino acid
a latent period and mitochondrial function is impaired
transport system L, which transports large amino acids19.
during the symptomatic period but not during the latent
It has been proposed that MeHg-cysteine conjugate phase. Although MeHg concentrations are maximal
is the pathway whereby MeHg exerts its toxicity20. Once during the latent period, the effects of MeHg on
NASCIMENTO et al: METHYLMERCURY NEUROTOXICITY 375

mitochondria could be indirect as they are preceded by including human fibroblasts 34, neuroblastoma, and
inhibition of protein synthesis28. Rats exposed to MeHg glioma cells36,37.
have reduced rates of cellular respiration and this effect
Neuroblastoma cells appear to be more susceptible
is reverted by the removal of K+, suggesting that there
to toxic effects of MeHg than either rat glioma or human
is an increase in K+ permeability of the mitochondrial
fibroblasts 38 , although the cell death mechanism
membrane29. High MeHg levels cause impairment of produced by MeHg in these cell lines has not been well
mitochondrial function as the organelle exhibits a characterized13.
membrane permeability transition state. MeHg exposure
induces a decrease in the activity of enzymes of the The integrity of microtubule function is critical for
mitochondrial energy metabolism such as cytochrome the physiological development of the CNS including
C oxidase (CCO), superoxide dismutase (SOD) and cell proliferation, migration of post-mitotic neurones
succinate dehydrogenase (SDH)28. This is probably due to form the cortical layers of the cerebrum and
to the decrease in the respiratory rate caused by MeHg- cerebellum, extension and stabilization of neuritis, and
induced inhibition of the tricarboxylic acid cycle. This axodendritic transport. Indeed, MeHg interference in
is consistent with previous work showing that MeHg microtubules is consistent with neuropathologic
exposure decreases succinate dehydrogenase activity30. findings in the autoptic brains of full-term newborn
In vitro MeHg exposure of isolated mitochondria from human infants from the Iraqi outbreak, who were
rat liver inhibits electron transport and phosphorylation, exposed in utero in early phases of pregnancy. These
increases K + permeability, and dissipates the findings included brain reduction, neuronal heterotopias
mitochondrial membrane potential (MMP)31. Loss of in the cerebral and cerebellar white matter, and abnormal
MMP results in efflux of mitochondrial Ca +2 and patterns of neuronal arrangement and alignment in the
inhibition of mitochondrial Ca+2 uptake32. In addition, cerebral cortex39.
MeHg exposure in isolated rat brain mitochondria The ability of MeHg in a concentration-dependent
causes ATP-dependent and -independent decrease in way to inhibit neuronal migration from the external
Ca +2 uptake and increase in Ca +2 efflux from granule layer towards the internal granule layer has been
mitochondria33. Although mitochondria participate in demonstrated experimentally in cerebellar organotypic
Ca+2 buffering at relatively elevated Ca+2, the affinity cultures40. It has been suggested that the extensive
of the uniport carrier for Ca+2 is low, and mitochondria apoptotic death observed in the external granule layer
may play only a minor role in buffering Ca2+ under following MeHg treatment is the result and not a cause
normal conditions32. of the impaired neuronal migration40.
Increased rate of Ca +2 is not observed during Interestingly, microtubule disruption provoked by
prolonged depolarization and it seems that under normal MeHg does not only affect the neuronal migration and
conditions mitochondria do not contain sufficient Ca2+ development of CNS. One of the most dangerous
to alter neurotransmitter release33. Recent evidence consequences of human exposure to relatively low
suggests that increased local concentration of Ca2+ may levels of MeHg is DNA damage as demonstrated by
be required for activation of neurotransmitter release. detection of micronucleated cells and chromosomal
Thus, the relatively small changes in Ca2+ observed upon aberrations40,41. In recent years, microtubule disruption
mitochondria depolarization are likely insufficient to was hypothesized as the origin of this genotoxic effect
alter the release of neurotransmitter via normal of mercury42-44 and a recent report demonstrated that
physiological mechanisms. However, other mechanisms very low MeHg concentrations are able to initiate
for MeHg toxicity could be trigger by small alterations genotoxic processes in human cell lines of brain origin45.
in the internal Ca2+ concentrations.
MeHg and neurotransmitters systems: Several metal
Microtubules network: MeHg seems to interact with compounds have been shown to interfere with
cytoplasmatic cytoskeletal components, including neurotransmission. MeHg directly affects the
microtubules34. In vitro studies demonstrated that MeHg mechanisms of neurotransmission, including release and
presents high affinity for tubulin sulphydryl groups uptake of neurotransmitters, enzymatic neurotransmitter
(-SH), depolymerizing cerebral microtubules and metabolic inactivation, and post-synaptic events
directly inhibiting their assembly 34,35. In addition, associated with receptor activation 46 . Some
several works reported that MeHg promotes neurotoxicants indirectly interfere with
microtubule disruption in a number of cell models, neurotransmission by interacting, for example, with
376 INDIAN J MED RES, OCTOBER 2008

energy metabolism, sodium channels, or ATPases. studied. In contrast, the chemistry of MeHg-MT
Furthermore, changes of any parameter of complexes has attracted much less attention. Earlier
neurotransmission can be the result of neuronal death reports demonstrated the inability of MT in the
due to cytotoxic effects of the neurotoxicants47. detoxification of MeHg and that it is unable to bind to
MeHg either in vivo or in vitro57,58. Subsequent attempts
The rising of extracellular glutamate levels is
to induce brain MT by exposure to MeHg + gave
responsible for the constant activation of metabotropic
inconsistent results: MT concentrations remained
and ionotropic glutamate receptors thus elevating Na+
unchanged in rats, whereas MT and mRNA
influx and Ca2+ release from intracellular organelles that
concentrations increased in MeHg-treated rat neonatal
may trigger a biochemical cascade which increases the
astrocyte cultures 58 . However, there is increasing
production of ROS47. Oxidative stress by itself inhibits
evidence that induction of MTs in astrocytes attenuates
the astrocytic glutamate uptake through a direct action
and even reverses the cytotoxicity caused by MeHg,
on the transporter proteins48,49.
indicating binding of MeHg by an astrocyte-specific
Although the toxic damage caused by MeHg might MT isoform, MT159.
be intrinsically prevalent in neurones, many of the
The MT association with heavy metals has been
published evidences suggest that neuronal damage in
the subject of several studies. For example, data about
response to MeHg most likely represents aberrant
geometry of mercury and metallothionein association
control of the extracellular milieu by the astrocytes50.
have demonstrated that characteristics of MeHg-MT
On line with this argument it should be remarked that
species cannot be extended to Hg-MT complexes57. The
the neurotoxic effect of MeHg could be reverted with
co-existence of digonal, trigonal-planar, and tetrahedral
antagonists of N-methyl-D-aspartate (NMDA)
co-ordination geometries together with the presence of
receptor48.
secondary mercury-sulphur interactions are common
Moreover, MeHg has been described to produce features in the chemistry of Hg(II) thiolates 57. In
increases in the spontaneous release of other contrast, MeHg shows a clear preference to form
neurotransmitters such as dopamine, GABA, acetylcholine, essentially linear two-coordinate Hg(II) complexes with
and serotonin from rat brain synaptosomes51-53. MeHg also thiolate ligands, even if, in some cases, secondary
inhibits astrocytic uptake of cystine and cysteine, the key interactions at the metal center are observed59,60. These
precursor for glutathione biosynthesis50. characteristics could reflect the differences in cellular
Methylmercury and metallothioneins: Metallothioneins resistances or response observed against mercury
(MTs) constitute a family of proteins characterized by organic and inorganic toxicity57.
an unusual cystein abundance54. Under physiological Methylmercury and oxidative stress
conditions, MTs are unusually rich in multiple cysteine
ROS are generally very small molecules and are
residues allowing their binding to metal centers and
highly reactive due to the presence of unpaired valence
enabling them to serve as a heavy-metal detoxification
shell electrons. ROS form as a natural by-product of
system 55. MTs are predominantly expressed in the
the normal oxygen metabolism and have important roles
central nervous system and it is important to gain new
in cell signalling. These molecules are generated
insight into how MTs are regulated in the brain in
continuously during oxidative metabolism and consist
pathological injury, such as that produced by MeHg
of inorganic molecules, such as superoxide radical anion
intoxication.
(O2–), hydrogen peroxide (H2O2), hydroxyl radicals
Some studies have reported the potential role of (OH–), as well as organic molecules such as alkoxyl
MTs in attenuating the cytotoxicity induced by and peroxyl radicals61. Some evidences suggest that the
MeHg54,56. Although the interaction of MTs with MeHg disturbance in the balance between oxidative and
ions has long been established, elucidation of the reductive cell processes is involved in the pathogenesis
binding features of MeHg-MT species has been of many neurodegenerative conditions such as
hampered by the inherent difficulties of MeHg-thiolate Alzheimer disease, amyotrophic lateral sclerosis (ALS),
chemistry, which mainly arise from the diverse co- and Parkinson disease. Other conditions such as
ordination preferences of Hg (II) and the various ligation autoimmune and inflammatory diseases, cancer, and
modes of the thiolate ligands 57 . Nevertheless, the diabetes mellitus also seemed to be related to this
analysis of MeHg binding to MTs has been intensively Disturbance61.
NASCIMENTO et al: METHYLMERCURY NEUROTOXICITY 377

MeHg has been thought to induce ROS and species69. The involvement of GSH in the neurotoxicity
generation of oxidative events leading to cell damage. of MeHg was also evaluated, showing that the increased
Previous studies have suggested that there is a oxidative stress is related with the depleted intracellular
relationship between these events with dysfunction of GSH levels 74,75 . The excessive formation of ROS
the cellular energetic metabolism and disruption of the induced by MeHg exposure can be reverted under
electron transport chain. These phenomena generate treatment with L-2-oxothiazolidine-4-carboxylic acid
oxidative stress62,63. MeHg exposure increases the rate (OTC), which increases the amount of intracellular
of ROS in the cerebellum (in vivo) and in the brain GSH, as well as the depletion of GSH by treatment with
synaptosomes as well as in the cerebellum neuronal buthionine-L-sulphoxane (BSO) can potentiate the
cultures, hypothalamic neuronal cell line, and mixed production of ROS induced by MeHg in rat primary
reaggregating cell cultures64-66. The formation of these cerebral astrocytes.
species was critical to determine the damage and the
Recently, a human population study in the Amazon
cell death in distinct cell types such as astrocytes and
correlated the MeHg exposure with the levels of
neurones.
glutathione and catalase activity. Surprisingly, it was
It seems that the intensity of MeHg exposure is a demonstrated that high blood levels of glutathione in
crucial factor to establish whether the neuronal death woman exposed to high concentrations of MeHg may
occurs by necrosis or apoptosis 67,68. However, the be explained by the increase of glutathione synthesis in
mechanism of cell death induced by oxidative stress response to oxidative stress or, more probably, by the
via MeHg has not been well characterized. inhibition of glutathione peroxidase activity77. In the
same population the inhibition of catalase activity was
Methylmercury and antioxidant defenses
also observed. These changes likely reflect adaptive
Many studies have already established that MeHg responses of the Amazonian population to oxidative
neurotoxicity evokes oxidative stress with formation stress induced by MeHg.
of ROS in the CNS and that the increase of ROS induces
Other studies revealed that the GSH content may
cell damage and death in the CNS. In order to avoid the
vary in different regions of the CNS, demonstrating that
damage caused by ROS, such as DNA strand breaks,
the GSH amount is higher in cerebral cells than in
lipid peroxidation, and protein modification,
cerebellar cells 78,79 . This may explain the higher
mechanisms have been developed during evolution
susceptibility of cerebellar cells to MeHg toxicity in
which dispose or prevent the generation of ROS69.
comparison with cerebral cells, but the reason why
However, the underlying mechanisms responsible for
certain areas of CNS showed different sensitivity to
the protection of CNS against MeHg neurotoxicity are
MeHg toxicity, remains unclear.
still poorly understood.
In addition, MeHg poisoning can induce
It is well known that cell defences against free
sympathetic ganglia toxicity and neurite outgrowth
radicals such as ROS include scavenger compounds
inhibition80,81. Compounds that possess sulphydryl (-SH)
such as glutathione, cysteine, melatonin, and enzymes
groups attenuate MeHg neurotoxicity, once at least part
with antioxidant activities as superoxide dismutase,
of MeHg effects occurs through interaction with -SH
catalase, and glutathione peroxidase70.
groups in cellular proteins82. In this context, primary
It was demonstrated that MeHg induces a neuronal cultures from avian sympathetic ganglion were
concentration-dependent increase in ROS formation in used to evaluate the protective role of antioxidants
rat neonatal neuronal culture and astrocytes agents with -SH group such as L-cysteine against MeHg
culture48,71,72. It was also shown that this effect can be toxicity. It was reported that MeHg induces massive
reverted by the use of n-propyl gallate (PG), a free cell death (neurite death) and that L-cysteine could fully
radical scavenger, superoxide dismutase (SOD), an protect (nearly 100%) the sympathetic neuron against
antioxidant enzyme, and α-phenyl-tert-butyl nitrone this damage. The effect of GSH was also tested showing
(PBN), a lipophilic hydroxyl radical spin trapping the same properties of cysteine83.
agent72,73.
The use of methionine, an antioxidant agent which
Endogenous glutathione (GSH) is one of the most does not possess -SH groups, fails to promote cell
abundant and essential thiol tripeptide present in protection against MeHg intoxication, proving the
mammalian cells for scavenging reactive oxygen relevance of -SH groups to this effect 83 . Another
378 INDIAN J MED RES, OCTOBER 2008

antioxidant that protects the brain against oxidative of MeHg produces significant increase in dopamine
stress is vitamin E, which maintains the integrity of release from rat striatum90. This increase is due to the
membrane by inhibiting lipid peroxidation84. Recent interaction of this metal with dopamine transporter
findings reported the protective effect of the (DAT). Pre-treatment with both GSH and cysteine
antioxidants tocopherols and tocotrienols (analogs to significantly decreased dopamine release induced by
vitamin E) against MeHg neurotoxicity85,86. In cerebellar MeHg, with GSH being more efficient than cysteine91.
granule cells (CGC), these compounds effectively
Concluding remarks
prevent cell death caused by MeHg intoxication as well
as improve cell migration87. The molecular mechanisms of MeHg damage in
both adult and developing CNS is not fully understood.
Evidences also suggest that the treatment with
Early reports have described a number of possible
trolox (6-hydroxy-2,5,7,8- tetramethylchroman-2-
cellular mechanisms to explain the neurotoxicity
carboxylic acid), other antioxidant derivate from
induced by MeHg. Most of these studies reported the
vitamin E, might provide prevention against oxidative
high affinity of MeHg for thiol groups (-SH) which are
stress. In MeHg-treated rats, it detected many apoptotic
present in cytoskeletal proteins, enzymes, and peptides
cells in the cerebellar granule layers and the treatment
that contain the amino acid cysteine78.
with trolox clearly repressed the appearance of these
apoptotic processes76. In accordance with these results, it is well
established that interactions between MeHg and -SH
A number of different hypotheses have been
potentially inactivate protein functions in all cellular
suggested to explain the mechanism by which the
and subcellular compartments42,92. The mechanisms
antioxidants defences protect CNS against MeHg
underlying the protective role of antioxidants molecules
neurotoxicity, which include scavenging and removal
against CNS toxicity by MeHg will likely be related to
of free radicals, reversal of glutamate uptake
their thiol groups (-SH) binding capacity.
impairment, inhibition of cytochrome c release, and
caspase activation88. Also in line with the above results, the susceptibility
of neurones to MeHg intoxication has been associated
It has been established that MeHg inhibits glutamate
to the absence or limited presence of inherent protective
transport by astrocytes by an unknown mechanism
mechanisms such as metallothioneins, reduced
which leads to the increase of ROS generation88. It was
glutathione, and other stress proteins93.
demonstrated that a variety of antioxidants can prevent
the overproduction of ROS, in this way attenuating This review corroborates with the major role of
MeHg neurotoxicity. Some workers have focused on ROS production in mediating MeHg toxicity in the CNS
the effect of antioxidant agents in the impairment of (Fig.). MeHg-induced toxicity with an emphasis on the
EAA transport elicited by MeHg. generation and role of reactive oxygen and nitrogen
species is reviewed. MeHg-mediated formation of free
In primary cultures of astrocytes from cerebral
radicals causes many modifications to DNA bases,
cortex, MeHg treatment inhibits the net uptake of 3H-
enhanced lipid peroxidation, and altered calcium and
D-aspartate, a glutamate analogue, that could be
sulphydryl homeostasis. Primary route for MeHg
completely prevented by catalase activity suggesting
toxicity is depletion of glutathione and bonding to
that the amount of H2O2 mediates this EAA transport
sulphydryl groups of proteins. Various studies have
inhibition89. The mechanisms whereby H2O2 alters EAA
confirmed that metals activate signalling pathways and
transport remains elusive, although it was proposed that
the cytotoxic effect of MeHg has been related to
the excessive production of H2O2 by MeHg
activation of mainly redoxsensitive transcription factors.
intoxication involves the inhibition of plasma membrane
Antioxidants (both enzymatic and non enzymatic)
Na+/K+-ATPase and subsequent modification in the ratio
provide protection against deleterious metal-mediated
of intracellular/extracellular Na + concentration
free radical attacks. Vitamin E and melatonin can
necessary for EAA transport. Interestingly, others
prevent the majority of MeHg-mediated damage both
antioxidants that prevent ROS formation such as trolox
in vitro systems and in metal loaded animals. Toxicity
and PBN did not present any effect in this transport
produced by MeHg showed that the protective effect
system89.
of vitamin E against lipid peroxidation may be
In previous studies it was demonstrated that associated rather with the level of non-enzymatic
intrastriatal administration of different concentrations antioxidants than the activity of enzymatic antioxidants.
NASCIMENTO et al: METHYLMERCURY NEUROTOXICITY 379

Fig. A schematic model of some of the currently proposed mechanism for cellular damage induced by MeHg in the CNS. In the extracellular
environment, MeHg inhibits glutamate uptake, as well as a number of the amino acids that are associated with the synthesis of astrocytic
glutathione (GSH). Accumulation of glutamate in the extracellular space and the resulting excessive activation of NMDA receptors can
result in excitotoxicity, and, ultimately, cell death. Other proposed mechanisms are related with mitochondrial MeHg -associated dysfunction,
including impaired cytoplasmatic Ca2+ homeostasis and release of ROS, metabolic inhibition that leads to impaired ATP production, lipidic
peroxidation and nuclear damage. MeHg also can provoke microtubules chain disruption decreasing vesicular migration or genotoxicity.

Molecular and cellular approaches can be a strategy 7. Harada M, Nakanishi J, Yasoda E, Pinheiro MC, Oikawa T,
to critically examine the possibility of therapeutic de Assis-Guimaraes G, et al. Mercury pollution in the Tapajos
River basin, Amazon: mercury level of head hair and health
actions such as antioxidants or chelanting agents in the effects. Environ Int 2001; 27 : 285-90.
treatment of neurodegeneration produced by MeHg.
8. Agarwal R, Kumar R, Behari JR. Mercury and lead content in
Acknowledgment fish species from the river Gomti, Lucknow, India, as
biomarkers of contamination. Bull Environ Contam Toxicol
This work was supported by JICA, CNPq #486351/2006-8, 2007; 78 : 118-22.
#620232/2004-8, CNPq-DCR/SEDECT #015/2004, and by the 9. Hunter D, Bomfond RR, Russell DS. Poisoning by
FINEP research grant “Rede Instituto Brasileiro de Neurociência methylmercury compounds. Q J Med 1940; 35 : 193-213.
(IBN-Net)” # 01.06.0842-00. LCLS and JLMN are CNPq research
fellows. 10. Hunter D, Russell DS. Focal cerebral and cerebellar atrophy
in a human subject due to organic mercury compounds.
References J Neurol Neurosurg Psychiatry 1954; 17 : 235-41.

1. Gochfeld M. Cases of mercury exposure, bioavailability, and 11. Steuerwald U, Weihe P, Jorgensen PJ, Bjerve K, Brock J,
absorption. Ecotoxicol Environ Saf 2003; 56 : 174-9. Heinzow B, et al. Maternal seafood diet, methylmercury
exposure and neonatal neurologic function. J Pediatr 2000;
2. Malm O. Gold mining as a source of mercury exposure in the 136 : 599-605.
Brazilian Amazon. Environ Res 1998; 77 : 73-8.
12. Cordier S, Garel M, Mandereau L, Morcel H, Doineau P,
3. Eisler R. Mercury hazards from gold mining to humans, plants, Gosme-Seguret S, et al. Neurodevelopmental investigations
and animals. Rev Environ Contam Toxicol 2004; 181 : 139- among methylmercury-exposed children in French Guiana.
98. Environ Res 2002; 89 : 1-11.
4. Clarkson TW, Magos L. The toxicology of mercury and its 13. Castoldi AF, Coccini T, Ceccatelli S, Manzo L. Neurotoxicity
chemical compounds. Crit Rev Toxicol 2006; 36 : 609-62. and molecular effects of methylmercury. Brain Res Bull 2001;
5. Eto K. Minamata disease. Neuropathology 2000; 20 : S14-9. 55 : 197-203.
6. Takeuchi T, Eto N, Eto K. Neuropathology of childhood cases 14. Lepham LW, Cermichiari E, Cox C, Meyers GJ, Baggs RB,
of methylmercury poisoning (Minamata disease) with Brewer R, et al. An analysis of autopsy brain tissue from infants
prolonged symptoms, with particular reference to the prematurely exposed to methylmercury, Neurotoxicology
decortication syndrome. Neurotoxicology 1979; 1 : 1-20. 1995; 16 : 689-704.
380 INDIAN J MED RES, OCTOBER 2008

15. Carta P, Flore C, Alinovi R, Ibba A, Tocco MG, Aru G, et al. dismutase prevents methylmercury toxicity in Hela cells. Life
Sub-clinical neurobehavioral abnormalities associated with Sci 1998; 62 : 157-61.
low level of mercury exposure through fish consumption. 31. Sone N, Larsstuvold MK, Kagawa Y. Effect of methyl mercury
Neurotoxicology 2003; 24 : 617-23. on phosphorylation, transport, and oxidation in mammalian
16. Silveira LCL, Damin ETB, Pinheiro MCN, Rodrigues AR, mitochondria. J Biochem 1977; 82 : 859-68.
Moura ALA, Côrtes MIT, et al. Visual dysfunction following 32. Levesque PC, Atchison WD. Disruption of brain mitochondrial
mercury exposure by breathing mercury vapour or by eating calcium sequestration by methylmercury. J Pharmacol Exp
mercury-contaminated food. In: Mollon JD, Pokorny J, Ther 1991; 256 : 236-42.
Knoblauch K, editors. Normal and defective colour vision.
Oxford, England: Oxford University Press; 2003. p. 407-17. 33. Denny MF, Hare ME, Atchison WD. Methylmercury alters
intrasynaptosomal endogenous polyvalent cation
17. Lakowicz JR, Anderson CJ. Permeability of lipid bilayers to concentrations. Toxicol Appl Pharmacol 1993; 122 : 222-32.
methylmercuric chloride: quantification by fluorescence
quenching of a carbazole-labeled phospholipid. Chem Biol 34. Sager PR, Doherty RA, Olmsted JB. Interaction of
Interact 1980; 30 : 309-23. methylmercury with microtubules in cultured cells and in vitro.
Exp Cell Res 1983; 146 : 127-37.
18. Sekine T, Cha SH, Endou H. The multispecific organic anion
transporter (OAT) family. Pflugers Arch 2000; 440 : 337-50. 35. Vogel DG, Margolis RL, Mottet NK. Analysis of
methylmercury binding sites on tubulin subunits and
19. Aschner M, Aschner JL. Mercury neurotoxicity: mechanisms microtubules. Pharmacol Toxicol 1989; 64 : 196-201.
of bloodbrain barrier transport. Neurosci Biobehav Rev 1990;
14 : 169-76. 36. Prasad KN, Nobles E, Ramanujam M. Differential sensitivity
of glioma cells and neuroblastoma cells to methylmercury
20. Morkrzan EM, Kerper LE, Ballatori N, Clarkson TW. toxicity in cultures. Environ Res 1979; 19 : 189-201.
Methylmercury-thiol uptake into cultured brain capillary
endothelial cells on amino acid system L. J Pharmacol Exp 37. Miura K, Inokawa M, Imura N. Effects of methylmercury and
Ther 1995; 272 : 1277-84. some metal ions on microtubule networks in mouse glioma
cells and in vitro tubulin polymerization. Toxicol Appl
21. Limke TL, Otero-Montanez JK, Atchison WD. Evidence for Pharmacol 1984; 73 : 218-31.
interactions between intracellular calcium stores during
methylmercury induced intracellular calcium dysregulation in 38. Sager PR, Syversen TLM. Differential responses to
methylmercury exposure and recovery in neuroblastoma and
rat cerebellar granule neurons. J Pharmacol Exp Ther 2003;
glioma cells and fibroblasts. Exp Neurol 1984; 85 : 371-82.
304 : 949-58.
39. Choi BH, Lapham LW, Amin-Zaki L, Saleem T. Abnormal
22. Marty MS, Atchison WD. Elevations of intracellular Ca+2 as a
neuronal migration, deranged cerebral cortical organization
probable contributor to decreased viability in cerebellar
and diffuse white matter astrocytosis of human fetal brain: A
granule cells following acute exposure to methylmercury.
major effect of methylmercury exposure in utero.
Toxicol Appl Pharmacol 1998; 150 : 98-105.
J Neuropathol Exp Neurol 1978; 37 : 719-33.
23. Limke TL, Heidemann SR, Atchison WD. Disruption of
40. Faustman EM, Ponce RA, Ou YC, Mendoza NA,
intraneuronal divalent cation regulation by methylmercury:
Lewandowski T, Kavanagh T, et al. Investigations of
are specific targets involved in altered neuronal development
methylmercury-induced alterations in neurogenesis. Environ
and cytotoxicity in methylmercury poisoning? Health Perspect 2002; 110 (Suppl 5) : 859-64.
Neurotoxicology 2004; 25 : 741-60.
41. Amorim MI, Mergler D, Bahia MO, Dubeau H, Miranda D,
24. Marty MS, Atchison WD. Pathways mediating Ca+2 entry in Lebel J, et al. Cytogenetic damage related to low levels of
rat cerebellar granule cells following in vitro exposure to methylmercury contamination in the Brazilian Amazon.
methyl mercury. Toxicol Appl Pharmacol 1997; 147 : 319-30. An Acad Bras Cienc 2000; 72 : 497-507.
25. Sakamoto M, Ikegami N, Nakano A. Protective effects of Ca2+ 42. Thier R, Bonacker D, Stoiber T, Böhm KJ, Wang M, Unger E,
channel blockers against methylmercury toxicity. Pharmacol et al. Interaction of metal salts with cytoskeletal motor protein
Toxicol 1996; 78 : 193-9. systems. Toxicol Lett 2003; 140 : 75-81.
26. Denny MF, Atchison WD. Elevations in the free 43. Bonacker D, Stoiber T, Wang M, Böhm KJ, Prots I, Unger E,
intrasynaptosomal concentration of endogenous zinc by et al. Genotoxicity of inorganic mercury salts based on
methylmercury. J Neurochem 1994; 14 : 290. disturbed microtubule function. Arch Toxicol 2004; 78 :
27. Mori N, Yasutake A, Hirayama K. Comparative study of 575-83.
activities in reactive oxygen species production/defense system 44. Stoiber T, Bonacker D, Böhm KJ, Bolt M, Their R, Degen G,
in mitochondria of rat brain and liver, and their susceptibility et al. Disturbed microtubule function and induction of
to methylmercury toxicity. Arch Toxicol 2007; 81 : 769-76. micronuclei by chelate complexes of mercury (II). Mutat Res
28. Yoshino Y, Mozai T, Nakao K. Biochemical changes in the 2004; 563 : 97-106.
brain in rats poisoned with an alkylmercury compound, with 45. Crespo-Lopez ME, Lima de Sa A, Herculano AM, Rodriguez
special reference to the inhibition of protein synthesis in brain Burbano R, do Nascimento JLM. Methylmercury genotoxicity:
cortex slices. J Neurochem 1966; 13 : 1223-30. a novel effect in human cell lines of the central nervous system.
29. Atchison WD, Hare MF. Mechanisms of methylmercury- Environ Int 2007; 33 : 141-6.
induced neurotoxicity. FASEB J 1994; 8 : 622-9. 46. Atchison WD. Is chemical neurotransmission altered
30. Naganuma A, Miura K, Tanaka-Kagawa T, Kitahara J, SekoY, specifically during methylmercury-induced cerebellar
Toyoda H, et al. Overexpression of manganese-superoxide dysfunction? Trends Pharmacol Sci 2005; 26 : 549-57.
NASCIMENTO et al: METHYLMERCURY NEUROTOXICITY 381

47. Orrenius S, Nicotera P. The calcium ion and cell death. 64. Ali SF, LeBel CP, Bondy SC. Reactive oxygen species
J Neural Transm 1994; 43 : 1-11. formation as a biomarker of methylmercury and trimethyltin
48. Park ST, Lim KT, Chung YT, Kim SU. Methylmercury induced neurotoxicity. Neurotoxicology1992; 13 : 637-48.
neurotoxicity in cerebral neuron culture is blocked by 65. Sarafian TA, Vartavarian L, Kane DJ, Bredesen DE, Verity
antioxidants and NMDA receptor antagonists. Neurotoxicology MA. bcl-2 expression decreases methyl mercury-induced free-
1996; 17 : 37-46. radical generation and cell killing in a neural cell line. Toxicol
49. Volterra A, Trotti D, Racagni G. Glutamate uptake is inhibited Lett 1994; 74 : 149-55.
by arachidonic acid and oxygen radicals via two distinct and 66. Sarafian TA. Methylmercury-induced generation of free
additive mechanisms. Mol Pharmacol 1994; 46 : 986-92. radicals: Biological implications. Met Ions Biol Syst 1999;
50. Shanker G, Allen JW, Mutkus LA, Aschner M. Methylmercury 36 : 415-44.
inhibits cysteine uptake in cultured primary astrocytes, but 67. Kunimoto M. Methylmercury induces apoptosis of rat
not in neurons. Brain Research 2001; 914 : 159-65. cerebellar neurons in primary culture. Biochem Biophys Res
51. Komulainen H, Tuomisto J. Interference of methylmercury Comm 1994; 204 : 310-7.
with monoamine uptake and release in rat brain synaptosomes. 68. Castoldi AF, Barni S, Turin I, Gandini C, Manzo L. Early
Acta Pharmacol Toxicol 1981; 48 : 214-22. acute necrosis, delayed apoptosis and cytoskeletal breakdown
52. Minema DJ, Cooper GP, Greeland RD. Effects of in cultured cerebellar granule neurons exposed to
methylmercury on neurotransmitters release from rat brain methylmercury. J Neurosci Res 2000; 60 : 775-87.
synaptosomes. Toxicol Appl Pharmacol 1989; 99 : 510-21.
69. Dringen R. Metabolism and functions of glutathione in brain.
53. Juárez BI, Martinéz ML, Montante M, Dufour E, García E, Prog Neurobiol 2000; 62 : 649-71.
Jiménez-Capdeville ME. Methylmercury increases glutamate
extracellular levels in frontal cortex of awake rats. Neurotoxicol 70. Olivieri G, Brack C, Muller-Spahn F, Stahelin HB, Herrmann
Teratol 2002; 24 : 767-71. M, Renard P, et al. Mercury induces cell cytotoxicity and
oxidative stress and increases beta-amyloid secretion and tau
54. Hidalgo J, Aschner M, Zatta P, Vasák M. Roles of the phosphorylation in SHSY5Y neuroblastoma cells.
metallothionein family of proteins in the central nervous J Neurochem 2000; 74 : 231-6.
system. Brain Res Bull 2001; 55 : 133-45.
71. Sorg O, Schilter B, Honegger P, Monnet-Tschudi F. Increased
55. González-Duarte P. Metallothioneins. In: McCleverty J, Meyer vulnerability of neurones and glial cells to low concentrations
TJ, editors. Comprehensive coordination chemistry II. of methylmercury in a prooxidant situation. Acta Neuropathol
Amsterdam: Elsevier-Pergamon; 2003; 8 : 213-28. 1998; 96 : 621-7.
56. Aschner M, Cherian MG, Klaassen CD, Palmiter RD, Erickson
72. Shanker G, Aschner M. Methylmercury-induced reactive
JC, Bush AI. Metallothioneins in brain: the role in physiology
oxygen species formation in neonatal cerebral astrocytic
and pathology. Toxicol Appl Pharmacol 1997; 142 : 229-42.
cultures is attenuated by antioxidants. Mol Brain Res 2003;
57. Wright JG, Natan MJ, MacDonnell FM, Ralston DM, 110 : 85-91.
O’Halloran TV. Mercury (II)-thiolate chemistry and the
mechanism of the heavy metal biosensor MerR. Prog Inorg 73. Gasso S, Cristofol RM, Selema G, Rosa R, Rodriguez-Farr'e
Chem 1990; 38 : 323-412. E, Sanfeliu C, et al. Antioxidant compounds and Ca2+ pathway
blockers differentially protect against methylmercury and
58. Rising L, Vitarella D, Kimelberg HK, Aschner M. mercuric chloride neurotoxicity, J Neurosci Res 2001; 66 :
Metallothionein induction in neonatal rat primary astrocyte 135-45.
cultures protects against methylmercury cytotoxicity.
J Neurochem 1995; 65 : 1562-8. 74. Lee YW, Ha MS, Kim YK. Role of reactive oxygen species
and glutathione in inorganic mercury-induced injury in human
59. Yao CP, Allen JW, Conklin DR, Aschner M. Transfection and
glioma cells. Neurochem Res 2001; 26 : 1187-93.
overexpression of metallothionein-I in neonatal rat primary
astrocyte cultures and in astrocytoma cells increases their 75. Shanker G, Syversen T, Aschner JL, Aschner M. Modulatory
resistance to methylmercury-induced cytotoxicity. Brain Res effect of glutathione status and antioxidants on methylmercury-
1999; 818 : 414-20. induced free radical formation in primary cultures of cerebral
60. Almagro X, Clegg W, Cucurull-Sánchez L, González-Duarte astrocytes. Mol Brain Res 2005; 137 : 11-22.
P, Traveria M. Schiff bases derived from mercury (II)- 76. Usuki F, Yasutake A, Umehara F, Tokunaga H, Matsumoto
aminothiolate complexes as metalloligands for transition M, Eto K, et al. In vivo protection of a water-soluble derivative
metals. J Organomet Chem 2001; 623 : 137-48. of vitamin E, Trolox, against methylmercury-intoxication in
61. Schulz JB, Lindenau J, Seyfried J, Dichgans J. Glutathione, the rat. Neurosci Lett 2001; 304 : 199-203.
oxidative stress and neurodegeneration. Eur J Biochem 2000; 77. Pinheiro MCM, Crespo-López ME, Vieira JLF, Oikawa T,
267 : 4904-11. Guimarães GA, Araújo CC, et al. Mercury pollution and
62. Clarkson TW. The toxicity of mercury. Crit Rev Clin Lab Sci childhood in Amazon riverside villages. Environ Int 2007;
1997; 34 : 369-403. 33 : 56-61.
63. Shanker G, Mutkus LA, Walker SJ, Aschner M. Methylmercury 78. Kaur P, Aschner M, Syversen T. Role of glutathione in
enhances arachidonic acid release and cytosolic phospholipase determining the differential sensitivity between the cortical
A2 expression in primary cultures of neonatal astrocytes. Brain and cerebellar regions towards mercury-induced oxidative
Res Mol Brain Res 2002; 106 : 1-11. stress. Toxicology 2007; 230 : 164-77.
382 INDIAN J MED RES, OCTOBER 2008

79. Adachi T, Kunimoto M. Acute cytotoxic effects of mercuric the natural vitamin E alphatocotrienol. J Neurochem 2006;
compounds in cultured astrocytes prepared from cerebral 98 : 1474-86.
hemisphere and cerebellum of newborn rats. Biol Pharm Bull 87. Shichiri M, Takanezawa Y, Uchida K, Tamai H, Arai H.
2005; 28 : 2308-11. Protection of cerebellar granule cells by tocopherols and
80. Soderstrom S, Ebendal T. In vitro toxicity of methyl mercury: tocotrienols against methylmercury toxicity. Brain Res 2007;
effects on nerve growth factor (NGF)-responsive neurons and 1182 : 106-15.
on NGF synthesis in fibroblasts. Toxicol Lett 1995; 75 : 133- 88. Aschner M, Syversen T, Souza DO, Rocha JBT, Farina M.
44. Involvement of glutamate and reactive oxygen species in
81. Miura K, Himeno S, Koide N, Imura N. Effects of methylmercury neurotoxicity. Braz J Med Biol Res 2007; 40 :
methylmercury and inorganic mercury on the growth of nerve 285-91.
fibers in cultured chick dorsal root ganglia. Tohoku J Exp Med 89. Allen JW, Mutkus LA, Aschner M. Methylmercury-mediated
2000; 192 : 195-210. inhibition of 3H-D-aspartate transport in cultured astrocytes
82. Mullaney KJ, Fehm MN, Vitarella D, Wagoner DE, Aschner is reversed by the antioxidant catalase. Brain Res 2001; 902 :
M. The role of -SH groups in methyl mercuric chloride-induced 92-100.
D-aspartate and rubidium release from rat primary astrocyte 90. Faro LRF, do Nascimento JLM, Alfonso M, Duran R.
cultures. Brain Res 1994; 641 : 1-9. Intrastriatal administration of methylmercury changes the in
83. Reis RAM, Herculano AM, Silva MCC, Santos RM, do vivo dopamine release from rat striatum. Neurochem Res 2000;
Nascimento JLM. In vitro toxicity induced by methylmercury 60 : 632-8.
on sympathetic neurons is reverted by L-cysteine or 91. Faro LRF, do Nascimento JLM, Campos F, Vidal L, Afonso
glutathione. Neurosci Res 2007; 58 : 278-84. M, Duran R. Protective effects of glutathione and cysteine on
84. Ricciarelli R, Zingg JM, Azzi A. Vitamin E: protective role of the methylmercuryinduced striatal dopamine release in vivo.
a Janus molecule. FASEB J 2001; 15 : 2314-25. Life Sci 2005; 77 : 444-51.
85. Osakada F, Hashino A, Kume T, Katsuki H, Kaneko S, Akaike 92. Gribble EJ, Hong SW, Faustman EM. The magnitude of
A. Alpha-tocotrienol provides the most potent neuroprotection methylmercuryinduced cytotoxicity and cell cycle arrest is p53-
among vitamin E analogs on cultured striatal neurons. dependent. Birth Defects Res A Clin Mol Teratol 2005; 73 :
Neuropharmacology 2004; 47 : 904-15. 29-38.
86. Khanna S, Roy S, Parinandi NL, Maurer M, Sen CK. 93. Sarafian TA, Bredesen DE, Verity MA. Cellular resistance to
Characterization of the potent neuroprotective properties of methylmercury. Neurotoxicology 1996; 17 : 27-36.

Reprint requests: Dr José Luiz Martins do Nascimento, Universidade Federal do Pará, Instituto de Ciências Biológicas
Laboratório de Neuroquimica Molecular e Celular, 66075-900 Belém, Pará, Brazil
e-mail: jlmn@ufpa.br

Potrebbero piacerti anche