You are on page 1of 37

C H A P T E R

45

Manganese
ROBERTO G. LUCCHINI, MICHAEL ASCHNER, YANGHO KIM, AND MARKO ŠARIĆ

ABSTRACT 1 INTRODUCTION

Manganese (Mn) is an essential element for As the fifth most abundant metal, manganese (Mn)
humans, animals, and plants. In humans and ani- is ubiquitous in the environment, comprising 0.1% of
mals the systemic Mn status is regulated by homeo- the Earth’s crust; soil levels are progressively increas-
static mechanisms. The natural presence of Mn and ing from anthropogenic sources. Mn is found in soil,
its compounds in the environment is being progres- air, deposited dust, water, and food. Its concentration
sively overridden by anthropogenic enrichment. in food varies markedly, but on the whole food is the
The main sources of Mn exposure are occupational, major source of Mn intake for humans.
including mining, iron/steel making, ferro/silico- Mn absorption by inhalation, ingestion, and via the
Mn alloy and dry alkaline battery production, and olfactory tract, and its distribution in the body, metab-
welding. Environmental Mn exposure is lower and is olism, and excretion are described based on findings
usually related to industrial emissions, the use of Mn from animal and human studies. The absorption of
compounds in agriculture and as fuel additives, and ingested Mn is regulated by homeostatic mechanisms
a high Mn content in drinking water. A high content and is predominantly achieved by regulation of its
of Mn in milk formula, in total parenteral nutrition excretion. Absorbed Mn is rapidly eliminated from
solution, and in drugs of abuse, as well as iron defi- the blood and first becomes concentrated in the liver,
ciency, can also increase the Mn body burden. Besides where it conjugates with the bile and is excreted into
through increased absorption, systemic Mn overload the intestine and feces. Urinary excretion is only about
can derive from impaired biliary excretion due to 0.01% per day of the body burden. The biological half-
hepatic insufficiency. The main toxic effects of Mn life in humans is 2-5 weeks, depending on body stores.
are on the central nervous system, including impair- Mn crosses the blood-brain barrier and accumulates in
ment of motor and cognitive function. Manganism is the brain, where the half-life is much longer than in
a neurological disorder that may occur in response blood.
to high inhalation exposures in occupational settings Mn is an essential element for humans, animals,
and which is characterized by mood changes and and plants. Although the daily Mn intake for many
an extrapyramidal syndrome resembling Parkinson people is likely to be below the estimated safe and
disease. Prolonged exposure to lower Mn doses can adequate levels, large-scale deficiency has not been
lead to an increased frequency of the signs of par- reported. Long-term exposure to increased concentra-
kinsonism. Effects on the respiratory, reproductive, tions of Mn, associated primarily with inhalation in
cardiovascular, hematological, endocrine, and immu- occupational settings, may result in neurological, neu-
nological systems are also discussed in this chapter, robehavioral, and mood effects. Research studies sug-
including genotoxicity and carcinogenicity. gest a direct link between chronic Mn exposures and

Handbook on the Toxicology of Metals 4E


http://dx.doi.org/10.1016/B978-0-444-59453-2.00045-7 975 Copyright © 2015 Elsevier B.V. All rights reserved.
976 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

parkinsonian-like disturbances. Mn-induced extrapy- 2004a; EPA, 2012a; ATSDR, 2012), drinking water
ramidal damage seems to occur postsynaptically to the guidelines (WHO, 2011; EPA, 2012b), and air quality
nigrostriatal system, predominantly in the globus pal- (WHO, 2000; EPA, 2010; Health Canada, 2010). Sev-
lidus and the striatum, as well as in other brain areas eral reviews have also been published (Bowman et al.,
such as the cortex. The principal mechanism of Mn 2011; Rivera-Mancía et al., 2011; Zheng et al., 2011;
neurotoxicity has yet to be clarified. Mn may poten- Guilarte, 2010; Lucchini et al., 2009; Yokel, 2009; Roth,
tiate the auto-oxidation or turnover of various intra- 2009; Aschner et al., 2009).
cellular catecholamines, such as dopamine, leading to
increased production of free radicals, reactive oxygen
species (ROS), and other cytotoxic metabolites. It may 2  PHYSICAL AND CHEMICAL PROPERTIES
disturb also γ-aminobutyric acid (GABA) regulation, as
well as glutamatergic neurotransmission. It also seems Manganese: atomic weight 54.94; atomic number
to impair cellular antioxidant defense mechanisms. 25; density 7.21-7.44 g/m3, depending on the allotropic
Lungs are the second major target organ affected by form; melting point 1245°C; boiling point 2097°C;
the exposure to Mn. Inhalation exposure to high concen- whitish-gray metal, harder than iron, very brittle. It
trations of Mn dioxide (or tetroxide) can cause chemi- is a transitional element that belongs to group VIIB
cal pneumonitis, an inflammatory response. Moreover, in the periodic table of the elements and it occurs in
an increased incidence of acute respiratory diseases— eight oxidation states, of which 0, II, IV, and VII are
bronchitis and pneumonia—has also been reported the most frequent, and valences I, III, V, and VI are
at low exposure levels, suggesting that inhaled Mn in rare. The only Mn cations are Mn2+ and Mn3+, which
an insoluble form can impair resistance to respiratory possess strong paramagnetic moments because of sev-
infections. This assumption has been supported by eral uncoupled electrons. Divalent Mn(II), present in
experimental and epidemiological studies. The influ- salts such as Mn chloride (MnCl2) or sulfate (MnSO4),
ence of Mn exposure has also been reported on the is the most stable valence; Mn dioxide (MnO2) is the
reproductive, cardiovascular, hematological, endocrine, most important Mn compound of the tetravalent state
and immunological systems. Although Mn can cause (IV), and potassium permanganate (KMnO4) of the
mutations, it is not deemed to be a carcinogenic metal. (VII) valence state. Most Mn salts are readily soluble in
Mn can be measured in various biological media. water, with only the phosphate and the carbonate hav-
Common biomarkers of exposure are blood and urine ing lower solubilities. Mn oxides are poorly soluble in
Mn. On a group basis, average levels of Mn in blood water. Mn forms various organometallic compounds
appear to be related to Mn body burden; in contrast, among which methylcyclopentadienyl Mn tricarbonyl
Mn in urine is less useful than blood-Mn (B-Mn) as an (MMT) is of major practical interest. Mn and its com-
exposure biomarker at the group level, because of the pounds can exist as solids in the soil and as solutes or
high degree of variability and relatively short biologi- small particles in water. Mn can also be present in fine
cal half-life (< 30 h). Thus, Mn in urine may to some and ultrafine dust particles in the air. Conversion fac-
extent reflect very recent exposure. On an individual tors: 1 μg Mn = 18.2 nmol Mn; 1 μg Mn/L ÷ 18.2 = 1 nmol
basis, blood and urine levels of Mn are not reliable Mn/L; 1  μg Mn/g creatinine = 2.06 nmol Mn/mmol
predictors of exposure to Mn. No significant correla- creatinine. The Chemical Abstracts Service (CAS) Nos
tion has been found between fecal excretion of Mn are: manganese 7439-96-5; Mn dioxide (Mn2O) 1313-
and exposure to the metal. Data on hair measurement 13-9; Mn oxide (Mn3O4) 1317-35-7; Mn chloride 7773-
in environmentally exposed children show a relation 01-5; Mn(II) sulfate 7785-87-7; Mn sulfate monohydrate
with the exposure over the previous months, and new 10034-96-5; potassium permanganate 7722-64-7; MMT
biomarkers like Mn in nails, saliva, or deciduous teeth, 12108-13-3.
or the Mn:Fe ratio in plasma or erythrocytes are being
assessed and deserve further validation.
The appearance of pallidal high signal intensities on 3  METHODS AND PROBLEMS OF
T1-weighted magnetic resonance imaging (MRI) may ANALYSIS
reflect a target organ dose of occupational Mn expo-
sure. In addition; Mn has a longer half-life in the brain Classical methods of analysis have been the peri-
than in blood. Thus, pallidal signal intensity is likely odate method for Mn in air, and the permanganate
to better reflect the cumulative dose compared to the method for Mn in water. However, these methods are
B-Mn level. not satisfactory for biological samples. Ideally, analysis
A number of documents on Mn are available by is now performed using inductively coupled plasma
international agencies regarding toxicity (WHO, 1986, atomic emission spectroscopy (ICP-AES). It is also
45 Manganese 977

possible to use flame atomic absorption spectroscopy may be digested in nitric acid prior to analysis. Sensi-
(AAS). In this method, a solution containing Mn is tivity is dependent on the volume of air drawn through
aspirated into a flame, and the concentration of Mn is the filter prior to analysis, and typical detection limits
determined from the light absorption at λ = 279.5 nm. are 1-2 μg/m3.
AAS has a lower sensitivity than ICP-AES and has gen- Standardization of the sampling criteria used for
erally been replaced by ICP-AES because of the greater measurement of airborne dust concentrations in the
suitability of ICP-AES for multielement analysis. High workplace has led to the development of particle size-
resolution ICP-coupled mass spectrometry (ICP-MS) selective sampling criteria for the inhalable PM (IPM),
has a greater sensitivity than ICP-AES. Progress in thoracic PM (TPM), and respirable PM (RPM) fractions
ICP-MS instrumentation was achieved by the intro- (CEN, 1993; ACGIH, 1999). Samplers are available for
duction of the collision cell interface to dissociate the collecting the inhalable fraction, for example the Insti-
many disturbing argon-based molecular ions, thermal- tute of Medicine (IOM) inhalable sampler (Mark and
ize the ions, and neutralize the disturbing argon ions Vincent, 1986). The Millipore 37-mm closed-face filter
of plasma gas (Ar+). Application of the collision cell in cassette (CFFC) sampler is still in use for the collection
ICP-MS results in higher ion transmission, improved of “total dust”; however, it does not comply with size-
sensitivity, and more precise isotope ratio measure- selective sampling criteria. Correction factors for IOM
ments compared to quadrupole ICP-MS without the and CFFC samples are generally between 1.0 and 2.5
collision cell (Becker and Dietze, 2000). (Tatum et al., 2001).
Neutron activation analysis is also an effective way Water may be analyzed directly. If the concentra-
to determine Mn concentrations in different samples tion of Mn is low, a concentration step (evaporation),
(Pejovic-Milic et al., 2009). This technique uses no extraction, and binding to a resin may be used. Detec-
reagents and a minimum of sample handling; thus, tion limits range from 0.005 to 50 μg/L. In all cases, acid
potential contamination with exogenous sources of Mn is added to the sample to prevent the precipitation of
can be avoided. It can be used for both environmental Mn. In a study performed by Beklemishev et al. (1997),
and biological samples. Other methods include spectro- a very low detection limit of 0.005 μg/L was obtained
photometry, MS, and X-ray fluorescence. Mn levels are for measuring Mn in tap and river water. Their ana-
nearly always reported as total Mn because none of the lytic method relied on an indicator reaction catalyzed
methods listed can distinguish between the different oxi- by Mn(II), which consists of the oxidation of 3,3′,5,5′-
dation states of Mn or between different Mn compounds. tetramethylbenzidine by potassium periodate (KIO4)
A simple determination of total Mn by atomic carried out on a paper-based sorbent surface.
absorption or gas chromatography followed by flame- Determination of Mn levels in soils, sludge, or other
ionization detection can be used to quantify MMT wastes requires an acid extraction/digestion step prior
concentrations in gasoline. By detecting the metal- to analysis. Treatment will usually involve heating in
lic portion of the compound, the detected MMT is nitric acid, oxidation with hydrogen peroxide, and filtra-
reported as Mn. Walton et al. (1991) described high tion and/or centrifugation to remove insoluble matter.
performance liquid chromatography coupled to laser-
excited atomic fluorescence spectrometry as a method
for detecting various species of MMT. 4  OCCURRENCE, PRODUCTION AND USES
As for other trace metal determinations, the analysis
of Mn in environmental media and in biological mate-
4.1  Occurrence and Production
rial where Mn is present should be performed with a
high standard of cleanliness to prevent external con- Mn is widely distributed in the Earth’s crust, com-
tamination. Analysis of Mn in blood may be influenced prising about 0.1% of the total; it is the twelfth most
by the anticoagulant used. A high concentration of Mn abundant element and the fifth most abundant metal
can be present in heparin. (ATSDR, 2012). Mn does not occur naturally in its
Mn in air exists as particulate matter (PM); therefore, native state as a base metal, but it is found in over 100
sampling is achieved by drawing air through a filter in minerals. Oxides, carbonates, and silicates are most
order to collect the suspended particles. A variety of important among Mn-containing minerals. The most
filter types (e.g. glass fibers and cellulose acetate) and common Mn mineral is pyrolusite (Mn dioxide), con-
sampling devices (low volume, high volume, personal, taining 60-63% Mn. Rhodocrosite (Mn carbonate) and
and dichotomous) are available, depending on the par- rhodonate (Mn silicate) are also common. Mn occurs in
ticle sizes of concern and the concentration range of nodules on the bottom of the ocean, in most iron ores,
interest. Material on the filter may be analyzed directly and over a wide range of concentrations as a natural
by X-ray fluorescence (Borgese et al., 2012), or the filter trace element in crude oil.
978 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

Most Mn is mined in open-pit or shallow under- porcelain and glass-bonding materials, and amethyst
ground mines. More than 80% of the high-grade Mn glass, and as the starting material for production of other
ore (> 35% Mn) is mined in South Africa, and also in Mn compounds. Manganese sulfate is used primarily as
Australia, Brazil, Gabon, India, and more recently a fertilizer and a livestock supplement where soils are
China and the former Commonwealth of Independent deficient in Mn, and in some glazes, varnishes, ceram-
States (CIS). According to the International Manga- ics, and fungicides. Potassium permanganate, due to its
nese Institute (IMnI, 2012), the world production of all strong oxidizing capacity, is used as a disinfectant, as an
grades of Mn ore (wet) decreased by 22% in 2009, to 36 antialgal agent, for metal cleaning, tanning, and bleach-
million metric tons (mt), which corresponds to 11 mil- ing, and as a preservative for fresh flowers and fruits,
lion mt in Mn units (content), a year-on-year decline and as well as in water and waste-treatment plants for
of 23%. In 2012, the global production of Mn ore (wet) water purification purposes (HSDB, 2012). Other uses
rose to 53 million mt, representing 16 million mt of Mn of Mn compounds are in textile bleaching, for linseed oil
units. China was the largest producer of Mn units (2.7 driers, in dyeing, in leather, tanning, and as an oxidizing
million mt), followed by South Africa (1.9 million mt), agent for the electrode coating of welding rods.
Australia (1.8 million mt), Brazil (1 million mt), Gabon Methcathinone (ephedrone), the oxidation prod-
(956,000 mt), India (845,000  mt), and Kazakhstan uct of ephedrine or pseudoephedrine, is a psycho-
(377,000 mt). Reserves of high-grade ore are about 500- stimulant that has become a substantial drug of abuse
600 × 106 tons, and those of low-grade ore are estimated (Kelly, 2011). Potassium permanganate can be used as
at several billion tons (Gerber et al., 2002). the oxidation agent, and Mn can therefore directly be
absorbed by drug users as an impurity in ephedrone.
Maneb and mancozeb are Mn-based fungicides.
4.2 Uses
Maneb is a manganous ethylene bis(dithiocarbamate)
Manganese is mainly used in metallurgical pro- sulfate or chloride, while mancozeb is a polymeric
cesses. About 90% of the global Mn production is used mixture of a zinc salt and maneb, containing 20% Mn
in steel manufacture as a deoxidizing and desulfur- and 2.5% zinc (HSDB, 2012). Among the organic Mn
izing additive, and as an alloy constituent. Most Mn compounds, MMT was developed in the 1950s as a
ore is smelted in electric furnaces to produce the ferro/ fuel additive to increase the octane level of gasoline
silico-Mn (Fe/Si-Mn) alloy widely used in the produc- and thus improve the antiknock properties of the fuel.
tion of steel. The production of 97-98% pure Mn metal It was introduced in Canada in 1976 and its use has
is achieved by aluminum reduction of low-iron-content increased so substantially that it completely replaced
Mn ore. Manganese with < 0.1% metallic impurities can tetraethyllead in gasoline in that country in 1990
be produced electrolytically from a Mn sulfate solution (Zayed et al., 1999). In 1977, MMT was banned by the
(HSDB, 2012). The total worldwide production of Mn U.S. Environmental Protection Agency (EPA, 1978) as
alloys dropped to 11.7 million mt in 2009, down 18% an additive in unleaded gasoline. In 1995, the ban was
from the previous year, but increased to 17.5 million mt lifted and a court decision ordered the EPA to again
by 2012. China continued to be the world’s largest Mn register the product for use as a fuel additive (EPA,
alloy-producing country, having produced nearly 6.6 1995), although testing for health effects continues.
million mt. It accounted for almost 57% of the global The Afton (former Ethyl) Corporation, the manufac-
output. The global economic recession caused a major turer of MMT, has been marketing its product to U.S.
drop in the demand for steel by 2009, which led to a refineries since late 1995 (Davis, 1999). Nevertheless,
decline in alloy production (IMnI, 2012); however, MMT is currently used only sparsely in the developed
3 years later, global steel production increased by 24%. world. The major refiners in Canada have voluntarily
Metallic Mn in the form of Fe/Si-Mn is principally stopped using MMT, owing to concern for the impact
used in steel production to improve hardness, stiff- of MMT on advanced vehicles, and as a result as much
ness, and strength. It is used in carbon steel, stainless as 95% of Canadian gasoline is now MMT free (Inside
steel, high-temperature steel, and tool steel, as well as in Fuels, 2004). MMT is not currently allowed in reformu-
cast iron and superalloys (HSDB, 2012). Manganese is lated gasoline, a gasoline blended with oxygenates to
a minor but indispensable component of welding con- reduce toxic pollutants primarily from older vehicles
sumables. Most consumables contain less than 6% Mn. (which is required in the USA in cities with the worst
The forms in which it is used include Fe-Mn, Si-Mn, and smog pollution) and is not used by the major oil com-
Mn carbonate. Mn compounds are produced either Mn panies in the United States. In Europe, MMT is used
from ores or Mn metal. Mn compounds have a variety in Greece (Geivanidis et al., 2003), in a couple of the
of uses. Manganese dioxide is commonly used in the Eastern countries, and perhaps by one small refiner
manufacture of dry-cell batteries, matches, fireworks, in Belgium (Blumberg and Walsh, 2004). Through
45 Manganese 979

Directive 2009/30/EC of the European Parliament and atmosphere, usually measured as total suspended par-
of the Council of 23 April 2009, EU Fuel Quality legis- ticulate Mn (TSP air-Mn). In areas with major foundry
lation set a limit of 6 mg Mn/L in gasoline on 28 April, facilities, annual averages of air-Mn may amount to
2012, falling to 2 mg/L as from 1 January, 2014. China 200-300 μg/m3 and Mn absorption from inhalation
is following the same requirements as Europe (Michael may rise to 4-6 μg/day. In areas associated with ferroal-
Walsh, personal communication). loy industries, it may be as high as 500 μg/m3 (intake
Another organic Mn compound, mangafodipir 10 μg/day, with the 24-h peak exceeding 200 μg/day)
[Mn(II) dipyridoxyl diphosphate (MnDPDP)], is clas- (WHO, 1981). Additional releases of Mn to the environ-
sified as a drug or therapeutic agent and used as both ment occur from natural sources (e.g. volcanos) and
a liver- and pancreas-specific contrast agent for MRI. from processes such as the combustion of cement.
It improves lesion detection of these organs in MRI by In recent years, much lower airborne Mn levels
selectively enhancing the normal parenchyma, but not have been reported for communities in the vicinity
the lesions, so that the contrast between tumorous and of active industrial facilities. In eight Mexican com-
normal tissues is increased (Wang et al., 1997). munities living at various distances from Mn-mining
operations, PM10 air-Mn levels averaged 420 ng/m3
(Rodriguez-Agudelo et al., 2006). In the surroundings
5  LEVELS AND FATE IN THE of a Brazilian ferro-Mn alloy factory, the PM2.5 air-Mn
ENVIRONMENT AND EXPOSURE levels averaged 150 ng/m3 (Menezes-Filho et al., 2011).
Within the framework of an epidemiological study in
Mn is ubiquitous in the environment. It occurs in Marietta, Ohio, the EPA used its AERMOD dispersion
airborne and deposited particles, water, soil, plants, model to estimate outdoor TSP air-Mn concentrations
and food. for individual study subjects; the mean estimated
long-term air-Mn exposure level was 180 ng/m3 (range
5.1  General Environment 40-960 ng/m3) (Kim et al., 2011a; Bowler et al., 2012).
Personal air sampling data were collected from 38
5.1.1  Ambient Air
children residing at a time-weighted distance (TWD)
Airborne Mn exists as a component of PM. A pop- between 4.7 and 28.5 km from the active ferroalloy
ulation-based study of personal exposure was carried plan in Marietta (Haynes et al., 2012). Mn concentra-
out in Riverside, California: participants were all non- tion in PM2.5 personal air samples ranged from 1.5
smokers above 10 years of age, and personal exposure to 54.5 ng/m3 [geometric mean (GM), 8.1]. TWD was
was assessed for Mn in the PM with a mean diameter a significant predictor of natural log personal air-Mn
of 10 μm (PM10) size fraction only (Clayton et al., 1993). concentration (lnMn) with an associated decrease of
Daytime exposures (median 49 ng/m3) were greater 0.075 lnMn for each km TWD (p< 0.05, 95% CI −0.13
than nighttime exposures (median 20 ng/m3). The larg- to −0.01). Personal PM10 air samples were recently col-
est population-based study of personal exposure to lected on 311 adolescents residing within 3 km from
airborne Mn was conducted in Toronto in 1995, dur- previously active ferroalloy plants in Valcamonica,
ing the widespread use of the MMT gasoline additive Italy (Lucchini et al., 2012). Mean (49.5 ng/m3), median
in Canada (Pellizzari et al., 1999). The mean personal (31.4 ng/m3), and range (1.24-517 ng/m3) levels were
PM2.5 Mn exposure was 9 ng/m3, with a 90th percentile significantly higher compared to a reference nonindus-
of 14 ng/m3. Median, mean, and 90th percentile of the trial area (27.4, 24.7, and 5.3-85.9 ng/m3, respectively).
personal PM10 Mn exposure were 15, 36, and 55 ng/m3 Compared to personal air sampling, data from
respectively. PM2.5 Mn made up a greater proportion of large-scale ambient monitoring programs allows the
PM10 Mn in indoor environments than in outdoor ones. spatial and temporal variability in air-Mn levels to be
Another population-based study was conducted in 1996 considered in both urban and rural settings. Annual
in Indianapolis, Indiana, a city without MMT use or averages of Mn in urban areas without significant Mn
major Mn-emitting industries at the time (Pellizzari et al., pollution are in the range of 0.01-0.07 μg/m3 (WHO,
2001). Median, mean, and 90th percentiles of the per- 2000). Based on measurements obtained in 102 U.S. cit-
sonal PM2.5 Mn exposure amounted to 3, 3, and 5 ng/m3 ies, the estimated background concentration is 40 ng/
respectively. Mn enrichment of personal samples (i.e. m3 (EPA, 2003; WHO, 2004a). Data compiled for 2006
mass of Mn per mass of PM) was higher in Toronto com- under the EPA Urban Air Toxics Monitoring Program,
pared to the U.S. locations (Health Canada, 2010). showed ambient air levels of Mn at 20 urban locations
Above average exposures to Mn are likely to across the United States. PM10 air-Mn was detected in
occur in or near factories, or at hazardous waste sites 415 samples of urban air at levels ranging from 0.24
that release significant amounts of Mn dust into the to 89.10 ng/m3 (EPA, 2007); the arithmetic mean, GM,
980 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

and median concentrations were 10.13, 6.68, and The rate of MMT degradation in natural aquifer and
6.29 ng/m3, respectively. TSP air-Mn levels ranged sediment systems is very slow under anaerobic condi-
from 0.85 to 614 ng/m3 in 114 air samples at the same tions. Calculated half-lives range from approximately
20 urban locations; the arithmetic mean, GM, and 0.2 to 1.5 years at 25°C (Garrison et al., 1995). However,
median concentrations of Mn in TSP were 47.89, 22.39, MMT photolyzes rapidly in purified, distilled water
and 23.98 ng/m3 respectively. exposed to sunlight (calculated half-life of 0.93 min).
Manganese can also be released into the air by In a survey of U.S. river waters (1974-1981), values
combustion of gasoline that contains MMT as an anti- ranging from < 11 μg/L to > 51 μg/L were reported,
knock additive. It has been estimated that if MMT with a median concentration of 24 μg/L for dissolved
were used in all gasoline, urban air-Mn levels would Mn (Smith et al., 1987). The average manganese con-
be increased by about 50 ng/m3 (Cooper, 1984). The centration in drinking water ranged from 4 to 32 μg/L
EPA (1994) estimated that a substantial number of (NAS, 1980; WHO, 1981). The EPA (2003) reported
people could be exposed to airborne Mn particulate median Mn concentrations in groundwater at 5 μg/L,
levels above 100 ng/m3 if 100% of gasoline contained with the 99th percentile at 2900 μg/L. In urban areas,
MMT. In Canada, where MMT in gasoline was already the median groundwater Mn concentration was found
widely in use, the estimated 10% per year increase in to be 150 μg/L, with the 99th percentile at 5600 μg/L.
Mn emission rates had been reported (Loranger and In public water systems supplied by groundwa-
Zayed, 1997; Zayed et al., 1999). Manganese can react ter, approximately 3% of the 982 sampled sources
with sulfur dioxide and nitrogen oxide, but the occur- exceeded the U.S. health reference level of 300 μg/L
rence of such reactions in the atmosphere has not been (EPA, 2003). According to the Geological Survey of
demonstrated. MMT is rapidly photolyzed by sun- Sweden (SGU, unpublished data), Mn concentrations
light in the atmosphere (half-time of less than 2 min) in Swedish groundwater used for drinking water are
(Garrison et al., 1995). on average 150 ± 510 μg/L (median 60  μg/L), with
maximum values as high as 30,000 μg/L. Around 20%
of the 12,000 sampled wells had Mn concentrations
5.1.2 Water
exceeding the Swedish recommended guideline value
Manganese may be released into water by discharge of 300 μg/L (Ljung and Vahter, 2007). High levels have
from industrial facilities or as leachate from landfills been reported in Bangladesh, where the average Mn
and soil. Concentrations of Mn in surface water are concentration in the wells was 800 μg/L, ranging from
usually reported as dissolved Mn. Some Mn com- 4 to 4000 μg/L (Wasserman et al., 2006).
pounds are readily soluble, so significant exposure can
also occur by the ingestion of contaminated drinking
5.1.3 Soil
water. However, Mn in surface water may oxidize or
adsorb to sediment particles and settle to the bottom. As a component of Earth’s crust, Mn occurs natu-
Manganese in soil can migrate as PM in air, as men- rally in virtually all soils. Mn enrichment in soil is
tioned before, but also in water, whereas soluble com- gradually increasing worldwide due to industrial
pounds may dissolve in water and leach from the soil. emissions (Herndon et al., 2011).
The extent of leaching is mainly determined by the The average natural (background) levels of Mn in
characteristics of the soil and is highly variable. The soils range from around 40 to 900 mg/kg, with an esti-
transport and partitioning of Mn in water is controlled mated mean background concentration of 330 mg/kg
by the solubility of the specific chemical form present, (Cooper, 1984). Surface soil Mn was measured within
which in turn is determined by pH, oxidation-reduc- 3 km of previously active ferroalloy plants in Valcamon-
tion potential, and the characteristics of the available ica, Italy (Lucchini et al., 2012). Average levels (mean
anions. The metal may exist in water in any of four 958 mg/kg, median 897, range 465-1729) were signifi-
oxidation states (II, IV, VI, VII). Manganese in water cantly higher compared to a reference nonindustrial
may be significantly bioconcentrated at lower trophic area (mean 427 mg/kg, median 409, range 160-734).
levels. A bioconcentration factor relates living organ- Significantly higher levels of Mn were also measured
isms to the concentration of the chemical in the water in lettuce grown from domestic gardens of the affected
in which they live. In order to protect consumers from area compared to the reference area (median 53.6 vs.
the risk of Mn bioconcentration in marine mollusks, 30.2 mg/kg) (Ferri et al., 2012).
the EPA has set a criterion for Mn at 0.1 mg/L for Accumulation of Mn in soil usually occurs in the
marine waters (EPA, 1993). Manganese in water may subsoil and not in the soil surface: 60-70% of Mn is
undergo oxidation at high pH, and it is also subject found in the sand fraction of the soil (WHO, 1981).
to microbial activity (Johnston and Kipphut, 1988). Soil Mn concentrations arising from the use of MMT
45 Manganese 981

in unleaded gasoline are apparently connected to Mn of the diet. Manganese intake can be higher in popu-
oxides from Mn-containing motor vehicle exhaust lations with high tea consumption. A cup of tea may
(Lytle et al., 1995). As they correlate with distance from contain 0.4-1.3 mg Mn (ATSDR, 2012).
the roadway, it can be concluded that the environmen- Considering all of the available data, it can be esti-
tal fate of Mn from vehicle MMT sources is associated mated that daily intake of Mn from food ranges from
with increased total Mn in soil (Normandin et al., 2.0 to 8.8 mg (WHO, 1981). Although gastrointestinal
1999). The oxidation state of Mn in soils and sediments (GI) absorption of Mn is low (3-5%), oral exposure is
may be altered by microbial activity (Ferri et al., 2012) the primary source for absorption of Mn in the general
population. Adequate and safe intake of Mn for adults
is considered to be 2-5 mg/day (NAS, 1980). According
5.2 Food
to some data from the USA, it is possible that a signifi-
Manganese is a natural component of most foods. cant proportion of Americans, especially females, are
Its concentration in foodstuffs varies markedly but, not consuming sufficient Mn (NAS, 1980; Pennington
on the whole, food constitutes the major source of Mn et al., 1986; Davis and Greger, 1992). However, infants
intake for humans. can ingest more than the estimated safe and adequate
The highest Mn concentrations are found in wheat dose of 0.3-1.0 mg/day for their age group (Penning-
and rice. Polished rice and wheat flour contain less Mn ton et al., 1986) because of relatively high Mn levels in
because most of it is in the bran. High concentrations prepared infant foods and formulas (Lönnerdal, 1997).
(up to 50 mg/kg) are found in nuts, tea, legumes, pine- The U.S. Food and Nutrition Board of the IOM has set
apples, and whole grains; lower levels (up to 5 mg/kg) adequate intake levels for Mn for both genders at dif-
are found in milk products, meat, fish, and eggs ferent life stages (FNB/IOM, 2001)
(Pennington et al., 1986; Davis, J.M., et al., 1992).
In connection with the levels of Mn in different
5.3  Work Environment
foodstuffs, it is interesting to quote a study on Mn
bioaccumulation by plants. Lytle et al. (1994) reported In the workplace, exposure to Mn occurs by inhala-
increased concentrations of Mn in the seeds and stems tion of airborne fumes and particles. This is a concern
of oats grown in the organic and sandy soils at the sta- mainly in the mining, Fe/Si-Mn, iron and steel, dry-cell
tion with the highest traffic density. In a study by Nor- battery, and welding industries (WHO, 1986). The aver-
mandin et al. (1999), Mn was measured in the plants age concentration of Mn in total dust was 0.210 mg/m3
(flower, stem, leaves, and root). The authors tried to in a study of South African mineworkers (Myers et al.,
evaluate the potential of dandelion (Taraxacum offici- 2003a). In a study on miners in the Middle East, aver-
nale) as a bioindicator of Mn arising from the use of age concentrations were between 62 and 114 mg/m3 of
MMT in gasoline. Mn concentrations of the different Mn in total dust, due to less effective control measures
parts of the plant did not correlate with the distance (Boojar and Goodarzi, 2002). For the ferroalloy indus-
from the roadways. In a Canadian study (Méranger try, the average inhalable Mn exposure measured in
and Smith, 1972), it was estimated that 54% of the Norway using IOM personal samplers for the inhal-
total Mn intake comes from cereals. The second larg- able fraction was 254 mg/m3, whereas the respirable
est source was potatoes, with 14%, whereas meat, fish, fraction averaged 0.028 mg/m3 (Ellingsen et al., 2003a).
and poultry provided only 2%. However, there is a dif- This study provided highly detailed exposure infor-
ference in Mn concentrations for the same items in dif- mation on inhalable Mn exposures, based on a total of
ferent countries and areas. In the USA and Germany, 153 side-by-side measurements. Information concern-
the average oral intake by adults has been estimated ing composition was grouped into four species, with
to range from 2.1 to 4.1 mg/day (Schelenz, 1977). In most of the Mn in the oxidation states 0 and 2+.
333 Korean subjects aged 19-87 years, 4.2 ± 2.3mg were Average exposure levels measured as Mn in total
reported (Bu and Choi, 2012). With a parallel collec- dust with the traditional CFFC samplers ranged from
tion of identical food aliquots (duplicate meal method) 0.005 to 1.49 mg/m3 (GM 0.054) among Italian ferroal-
and chemical analysis, the average intake amounted loy workers (Lucchini et al., 1999). Among welders,
to 2.88 mg/day (standard deviation 0.64) (Nkwenkeu Mn concentrations can vary from 0.1 mg/m3 (Kucera
et al., 2002). et al., 2001) to 1.5 mg/m3 (Lu et al., 2005) and 4 mg/m3
Variations in Mn intake can to a large extent be (Korczynski, 2000), with peak exposure levels more
explained by differences in nutritional habits. In frequently reached in confined spaces in the shipbuild-
populations with cereals and rice as the main food ing industry. The GM concentrations of Mn and iron
sources, intake will be higher compared to areas among welders were 97 μg/m3 (range 3-4620, n = 188)
where meat and dairy products make up a larger part and 894 μg/m3 (range 106-20,300; n = 188), respectively
982 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

(Ellingsen et al., 2006). No substantial difference was 6 TOXICOKINETICS


observed in the air-Mn concentrations when welding
mild steel compared to welding stainless steel. Personal 6.1 Absorption
sampling of welding fumes was recently carried out
in 241 German welders during a working shift. Respi- 6.1.1 Inhalation
rable Mn was measured, with a median of 62 μg/m3 There are no quantitative animal data on absorption
(interquartile range 8.4-320  μg/m3); inhalable Mn rates for inhaled Mn. As Mn dioxide and other inhaled
amounted to similar concentrations (median 73 μg/m3; Mn compounds are practically insoluble in water, only
interquartile range 10-340 μg/m3) (Pesch et al., 2012). Mn in particles small enough to reach the alveolar lining
These data show that Mn particulate mainly occurs can be absorbed into the blood (WHO, 1981). However,
in the respirable fraction for welding fumes. Man- particles that are deposited in the upper airways may
ganese exposure among welders is often in excess of be moved by mucociliary transport to the throat, where
the American Conference of Governmental Industrial they are swallowed and enter the stomach; thus, Mn
Hygienists’ (ACGIH, 1995) threshold limit value (TLV) in particulate becomes available for GI absorption (see
of 0.2 mg Mn/m3 for the inhalable fraction (Flynn and Section 6.1.2). This process has been found to account
Susi, 2010). Considering the neurotoxic effects of Mn on for clearance of a significant fraction of Mn-containing
“CNS (i.e. central nervous system) function (neurobe- particles initially deposited in the lung (Drown et al.,
havioral and neuropsychological changes),” in 2013 1986; Newland et al., 1987). In a study performed by
the ACGIH adopted a lower TLV of 20 μg/m3 for the Tjälve et al. (1996), the uptake of Mn into brain regions
respirable fraction (ACGIH, 2013). Based on findings of weanling male Sprague-Dawley rats following intra-
in epidemiological studies and a recent meta-analysis nasal administration of 4 μg/kg 54Mn was investigated.
(Meyer-Baron et al., 2009) showing a reduced perfor- Whole-body autoradiography of the rats at different
mance of Mn-exposed workers, predominantly in tests time points revealed that the olfactory bulb contained
associated with motor functions, the German MAK the vast majority of measured Mn at 1, 3, and 7 days
commission has recommended 200 μg/m3 for inhalable postdosing (90%, 69%, and 47%, respectively), with val-
Mn and 20 μg/m3 for respirable Mn as MAK values. ues decreasing to a low of 16% at 12 weeks. Significant
The EU Scientific Committee on Occupational Expo- uptake of Mn by other brain regions was not observed
sure Limits (SCOEL) recommends a 8-h time-weighted until the third day, when the basal forebrain, cerebral
average (TWA) of 200 μg/m3 for the inhalable fraction cortex, hypothalamus, and striatum contained 21%, 2%,
and 50 μg/m3 for the respirable fraction (SCOEL, 2011). 3%, and 1% of the measured label, respectively. Maxi-
Assuming a TWA level of 1 mg/m3, which is the mum uptake of the label by these regions was observed
National Institute for Occupational Safety and Health at 7 days postdosing, with the percentage of the label
(NIOSH, 2005) recommended exposure limit, the maxi- in the basal forebrain reaching 28%, and with the other
mum Mn inhalation exposure would be 10 mg Mn/day parts containing amounts slightly more than double
(assuming inhalation of 10 m3 air during an 8-h work- their 3-day values. The liver and kidneys each contained
day). The study by Roels et al. (1992) in alkaline battery approximately 1% of the measured Mn at 1, 3, and 7
workers reported a 5% probability of increased hand days, with values decreasing consistently to 12 weeks.
tremor for a time-integrated air-Mn exposure thresh- Roels et al. (1997) measured B-Mn levels in
old of 3.575 mg Mn/m3 × year (total dust) and 0.730 mg 3-month-old rats following endotracheal adminis-
Mn/m3 × year (respirable dust), and suggested an 8-h tration of 1.22 mg Mn/kg as either MnCl2 or MnO2.
TWA of 0.089 mg/m3 for total dust and 0.018 mg B-Mn following MnCl2 (a water-soluble compound)
Mn/m3 for respirable dust as a protective level to administration reached a maximum 30 min postdos-
prevent the onset of early psychomotor effects. The ing, whereas with MnO2 (insoluble compound) , the
lowest observable adverse effect level (LOAEL) maximal B-Mn level was reached with a much slower
for total dust derived by Lucchini et al. (1999) was rate 168 h postdosing. The authors also studied the
0.10 mg Mn/m3 based on neurobehavioral changes. distribution of Mn in blood, liver, and brain regions
Mergler et al. (1994) calculated a LOAEL of 0.035 mg/m3 (cortex, cerebellum, striatum) following endotracheal
for the respirable fraction, based on early signs of administration of these Mn compounds (once a week,
neurotoxicity in workers of a Fe/Si-Mn alloy plant. 4 weeks). Compared to control rats, this subchronic
As far as occupational exposure to Mn from the com- study of pulmonary Mn exposure showed a signifi-
bustion of MMT is concerned, in a study performed cant increase in B-Mn, no increase in hepatic Mn, and
by Sierra et al. (1995) in Montreal, Canada, it was significantly increased Mn deposition in the differ-
shown that less than 10% of the Mn exposure of ent brain regions, with a selective increase of striatal
garage mechanics was due to MMT. Mn concentration (205%) for the more bioavailable
45 Manganese 983

MnCl2. Other studies in rats indicated that absorption The uptake of Mn is decreased by preexposure to high
via the olfactory mucosa is a potential contributor to dietary levels of Mn (Davis, C.D., et al., 1992).
brain Mn deposition. Since such a route would bypass Studies performed in suckling rat pups (Lönnerdal,
liver uptake and biliary excretion of Mn, its transport 1987, 1997) demonstrated different absorption rates of
by the olfactory nerve directly to the brain may have Mn for different milks and formulas. Infants fed cow
toxicological importance (Fechter, 1999). The trans- milk formula may retain five times more Mn, and
port of Mn across the olfactory pathway is further infants fed soy formula may retain 25 times more Mn
demonstrated for ultrafine particles and may result in than breast-fed infants.
the accumulation of Mn and inflammatory changes at Iron deficiency increases the GI uptake of Mn (Kim
the olfactory bulb to a greater extent than in the lung and Lee, 2011). As dietary iron (Fe) intake appears to
(Elder et al., 2006). be one of the key determinants of GI absorption of
The primary combustion products of MMT are Mn Mn, their interdependence has been extensively inves-
phosphate, Mn sulfate, and a Mn phosphate/sulfate tigated. The interdependence of iron and Mn may be
mixture. Salehi et al. (2003) carried out a study on 129 due to the fact that iron and Mn are absorbed by the
6-week-old male Sprague-Dawley rats divided in four same transport system [natural resistance-associated
groups—one as control and three groups exposed to a macrophage protein 2/divalent metal transporter 1
Mn phosphate/sulfate mixture in inhalation chambers (DMT-1)] in the gut (Rehnberg et al., 1982). Both Mn2+
for a period of 13 weeks (5 days a week, 6 h a day). Expo- and Fe2+ are bound by serotransferrin (Tf) and com-
sure concentrations were 3000, 300, and 30 μg Mn/m3. At pete for binding to this protein in the body. Therefore,
the end of the exposure period, locomotor activity and diets that are low in iron allow Tf to bind more Mn2+.
resting time tests were conducted for 36 h using a com- Interaction between iron and Mn occurs only between
puterized autotrack system. Rats were then euthanized nonheme iron and Mn (Davis, C.D., et al., 1992).
by exsanguinations and Mn concentrations in different Manganese interactions with other elements (Cd,
tissues, blood, and brain were determined by neutron Ni, In, Rh, and Se) at the level of GI absorption were
activation analysis. Increased Mn concentrations were also observed (Jacobs et al., 1978). High dietary intakes
found in the blood, kidney, lung, testes, and all brain sec- of phosphorus (Wedekind et al., 1991) can decrease Mn
tions in the highest exposure group. Manganese in the uptake in chicks. More recent data also confirm that
lung and the olfactory bulb was dose dependent. Rats the absorption of Mn is inversely related to the level of
exposed to 300 and 3000 μg Mn/m3 exhibited significant calcium in the diet (Lutz et al., 1993).
body-weight loss in comparison with the control group. Manganese absorption is age-dependent. Dorner
et al. (1989) have shown that infants, especially pre-
6.1.2 Ingestion
mature infants, retain a higher proportion of Mn than
The amount of Mn absorbed across the GI tract in adults. Animal studies support these findings (Rehn-
animals and humans depends mainly of the amount berg et al., 1980, 1982, 1985). Such age-dependent dif-
ingested and the iron status. In the absence of iron defi- ferences in the retention of Mn could also be due to
ciency, GI absorption in humans averages about 3-5% differences in excretory ability (Miller et al., 1975), or
(Davidsson et al., 1988, 1989). to age-related changes in the dietary intake levels of
In animal studies it was shown that Mn is absorbed iron and Mn (Ballatori et al., 1987). Animal studies
equally well throughout the length of the small intes- show that absorption and/or retention of Mn is higher
tine (Thomson et al., 1971). On the other hand, Miller in neonates, but returns to the level of older animals
et al. (1972) found in calves that the upper sections of at approximately postgestational day 17-18 (Rehnberg
the small intestine absorb far more 54Mn than the lower et al., 1982; Lönnerdal, 1987). Dorner et al. (1989) do
section. It seems that there is not a marked difference not provide adequate data to determine when this
between the retention of Mn ingested via food or water transition takes place in human infants. In a more
(Davidsson et al., 1988, 1989), but the absorption rate recent study by Kostial et al. (2005) on the regulation of
depends on the water solubility of the Mn compounds Mn in perinatally exposed rat pups, it was shown that
(Johnson et al., 1991; EPA, 1995). the concentration of Mn in these pups (whose mothers
Studies of oral absorption of Mn in animals have were orally exposed to Mn in drinking water—as Mn
yielded results that are generally similar to those in chloride at a dose of 2000 mg Mn/L throughout preg-
humans. In pigs, which have GI tracts similar to those nancy and 11 days of lactation) was six to eight times
of humans, Mn uptake was measured using orally higher than in controls, irrespective of the period and
administered labeled Mn. The results showed a mean duration of exposure. After cessation of exposure, the
absorption rate of 5% 1-6 h postdosing, 7% 6-12 h post- Mn concentration decreased almost to control levels.
dosing, and 3.8% 12-24 h postdosing (Finley et al., 1997). These results indicate the highly efficient regulation of
984 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

Mn accumulation in pups exposed to Mn during the examined, providing an alternative explanation for
perinatal period. There are no studies on the absorp- the efflux of Mn from the brain (Aschner, 2006a,b).
tion of Mn following oral exposure to MMT in either Vitarella et al. (2000) exposed adult rats to airborne
humans or animals. doses of particulate Mn, as Mn phosphate, at 0, 0.03,
0.3, 3 mg/Mn/m3. The particles had a mean diam-
6.1.3  Dermal Exposure eter of 1.5 μm. Exposure lasted for 6 h/day for either 5
days/week (10 exposures) or 7 days/week (14 expo-
Although many forms of manganese are water sol-
sures). Plasma, erythrocytes, olfactory bulb, striatum,
uble, there is little evidence that dermal contact with
cerebellum, lung, liver, femur, and skeletal muscle
manganese results in significant absorption through
were analyzed for Mn content using neutron activation
the skin. Occupational exposure to MMT may be sig-
analysis following exposure to 3 mg/m3 (after either
nificant via dermal absorption for gasoline attendants
dosing regimen). A lower dose of 0.3 mg/m3 resulted in
or mechanics (ATSDR, 2012).
increased Mn concentrations in the olfactory bulb and
lung (14-dose regimen only). Striatal Mn levels were
6.2 Distribution
increased at the two highest doses only after 14 days of
Manganese is a normal component of human and exposure. Concentrations in the cerebellum were simi-
animal tissues and fluids, with the highest levels in larly elevated, which was interpreted by the authors to
the liver, pancreas, and kidney and the lowest levels in indicate that accumulation of Mn was not selective for
bone and fat. Most tissue concentrations range between the striatum. Red blood cell and plasma Mn levels were
0.1 and 1 μg Mn/g wet weight (Sumino et al., 1975). increased only in rats exposed to the highest dose for
Absorbed Mn is rapidly eliminated from the blood the 10-day exposure period. These data indicate that
and first concentrates in the liver. Excess metal may Mn can accumulate in the olfactory bulb even at lower
be distributed to other tissues. Manganese is distrib- doses. However, it must be noted that tissues were col-
uted throughout the body at relatively constant con- lected and analyzed directly after cessation of exposure.
centrations that are characteristic of the individual It is not clear whether the distribution of Mn would be
tissues and almost independent of the species. Gen- similar if tissues had been analyzed at a later time.
erally, organs and tissues do not accumulate large Studies on humans suffering from chronic liver dis-
concentrations of Mn. Minor amounts go to the brain ease or some other liver dysfunction (e.g. cirrhosis, por-
and bone, as shown in experiments on mice and tacaval shunt) following oral exposure, presumably via
monkeys (Dastur et al., 1971). The brain has a small the diet, with impaired Mn excretion showed that Mn
amount of Mn, but its retention time is rather long. preferentially accumulates in the basal ganglia, espe-
In monkeys, it was shown that elimination from the cially the globus pallidus and the substantia nigra. Tl-
cerebrum was very slow. While the half-time in the weighted MRI or neutron activation analysis were used
whole body was estimated to be 95 days for the slow to determine the accumulation of Mn (Devenyi et al.,
component, it was not possible to obtain an estimate 1994; Hauser et al., 1996; Fell et al., 1996; Pomier-Layr-
for the cerebrum even after 278 days, indicating argues et al., 1998; Rose et al., 1999; Lucchini et al., 2000).
an extremely long biological half-time in this tissue The distribution of Mn in the brain was investigated
(Dastur et al., 1971). In 1999, Aschner wrote a review using capuchin (Newland et al., 1989; Newland and
on Mn homeostasis in the CNS, focusing on the mech- Weiss, 1992) and macaque (Newland et al., 1989) mon-
anisms of transport across the blood-brain barrier, keys given intravenous injections of MnCl2 that reached
and particularly on transport mechanisms associated a cumulative dose of 10-40 mg Mn/kg. MRI indicated
with the regulation of Mn uptake and release by astro- a symmetric hyperintensity in the globus pallidus and
cytes and its requirement for glutamine synthetase substantia nigra consistent with accumulation of Mn in
function and regulation (Aschner, 1999). Transport these areas. A substantial accumulation of Mn was also
across the blood-brain barrier was recently attributed noted in the pituitary at low cumulative doses.
to an active carrier-mediated mechanism for influx Some experimental animal studies showed that
into the brain and to a passive diffusive mechanism large increases in tissue levels of Mn compared to the
for efflux from the brain (Yokel and Crossgrove, 2004; controls occurred in rats over the first 24 days of expo-
Crossgrove and Yokel, 2005). This would be an impor- sure to 214 mg Mn/kg body weight/day (as Mn3O4)
tant mechanism to explain the occurrence of a brain for up to 224 days. However, levels tended to decrease
overload after excessive absorption of Mn, but also toward the control levels as exposure was continued
after prolonged exposure to low doses. In a review (Rehnberg et al., 1980; Kristensson et al., 1986). This pat-
on the transport of Mn across the blood-brain barrier, tern is thought to be due to a homeostatic mechanism
putative carriers for Mn into and out of the brain were that leads to decreased absorption and/or increased
45 Manganese 985

excretion of Mn when Mn intake levels are high (Bal- to Tf and in the bivalent state to an α-macroglobulin
latori et al., 1987). Davis, C.D., et al. (1992) and Malecki (Gibbons et al., 1976). The plasma content of Mn is much
et al. (1996) demonstrated that rats fed elevated lev- lower than the Mn content of red blood cells (1:5 ratio)
els of Mn for several weeks had increased tissue Mn (Weissman, 1981). In healthy control adults on a normal
concentrations, despite increased endogenous losses of diet, about 66% of Mn in the blood is found in red blood
Mn from the gut as biliary Mn. cells, 30% in white blood cells and platelets, and only 4%
Manganese penetrates the placental barrier in all in the plasma (Milne et al., 1990).
species. Children are exposed in utero because Mn in The organic Mn compound MMT is rapidly metabo-
maternal blood crosses the placenta to satisfy the fetal lized in rats. After oral exposure and injection, the liver,
need for Mn. Manganese has been measured in cord kidneys, and lungs contained the highest concentra-
blood plasma of premature and full-term infants and tions. The distribution in general was similar to that seen
their mothers (Wilson et al., 1991). In a human study on after exposure to inorganic Mn. Gianutsos et al. (1985)
Mn levels during pregnancy and at birth performed in administered 0, 11, or 22 mg Mn/kg as MMT (dissolved
a Southwest Quebec population (Takser et al., 2004), it in propylene glycol) to male adult mice via subcutane-
was shown that maternal B-Mn levels increased signif- ous (s.c.) injection. The experiment was divided into an
icantly during pregnancy and cord B-Mn levels were acute study (one dose) and a “chronic” study (10 doses),
significantly higher than those of maternal blood. This and accumulation of Mn in the brain was followed. The
study also indicated that lifestyle (mother’s smoking brain Mn level 24 h after the single dose of MMT was
habit) and environmental factors might interfere with significantly higher than control values: after 10 doses of
the balance and homeostatic mechanisms required to 11 mg/kg or 22 mg/kg, the brain Mn levels were signifi-
maintain Mn at optimal levels for the physiological cantly higher than the control values, and significantly
changes that occur during pregnancy. In a previous different than the levels reported after acute exposure.
study, levels of Mn in the umbilical cord and in mater- Manganese levels in the brains of mice given a single
nal blood were compared between 160 mother-neonate dose of MMT at 22 mg Mn/kg were also compared with
pairs in Montreal and 206 pairs in Paris. The prevalence those following injection of the same Mn dose as MnCl2.
of high Mn levels in umbilical cord blood was found to The Mn level in brain after MnCl2 exposure followed
be significantly higher in Montreal, where MMT was the same increasing trend over 24-h analysis as seen fol-
in use in gasoline in 1977 (Smargiassi et al., 2002). lowing the MMT application, but was somewhat higher
Manganese is secreted into milk. Its concentration at each time point, with a maximum value of > 2.0 μg/g
in breast milk varies between 3.4 and 10 μg/L (Collipp wet weight (compared with ∼1.4 μg/g wet weight
et al., 1983; Arnaud and Favier, 1995). following MMT exposure).
Manganese is more uniformly distributed in fetal Another study included young adult rats admin-
than in adult tissues. A study on age-related reten- istered with MMT dissolved in propylene glycol via
tion and distribution of ingested Mn3O4 in rats (Cahill s.c. injection at a dose of 1 mg Mn/kg (McGinley et al.,
et al., 1980) demonstrated that 24 h after ingestion 1987). The rats were sacrificed 1.5, 3, 6, 12, 24, 48, or 96 h
infant rats retained up to 20 times more Mn compared postinjection. The maximum accumulation of MMT-
to adolescent or adult rats. The retention of Mn was derived Mn measured 3 h after dosing was found pri-
also a function of age at exposure in infancy. The liver marily in the lungs (∼9 mg/kg), kidneys (3.9 mg/kg),
was the principal site of deposition at all ages. Brain liver (2.75  mg/kg), and blood (∼0.75 mg/kg). The
concentrations of Mn from a single dose were easily concentrations of Mn in these four tissues were still
detectable in infants but not in adults. A study measur- elevated (∼1 mg/kg) at 96 h postdosing. Brain Mn con-
ing the retention of a single oral dose of radiolabeled centrations were not significantly elevated over con-
Mn in adult and neonatal rats indicated that retention trol levels in MMT-treated animals.
of the label 6 days after exposure was much greater In connection with the clinical use of mangafodipir
in pups. The addition of Mn to the animals’ drinking as a contrast agent, several qualitative studies have
water decreased retention of the radiolabel in pups shown the presence of Mn in the liver due to increased
and adults (Kostial, 1989). A high rate of Mn absorp- Tl-weighted MRI signal in that organ following man-
tion and distribution in selected tissues, especially the gafodipir administration at 0.17-0.83  mg Mn/kg
cerebrum, hypothalamus, and pituitary, was found in (Bernardino et al., 1992; Lim et al., 1991; Padovani et al.,
preweanling rats dosed daily with particulate Mn3O4 1996; Wang et al., 1997). Several studies (Gallez et al.,
for 12 or 27 days postpartum (Rehnberg et al., 1982). 1997; Hustvedt et al., 1997; Grant et al., 1997) have
After GI or pulmonary absorption, Mn is transported determined the distribution of Mn in tissues of ani-
in the blood bound to proteins and low molecular weight mals following the intravenous (i.v.) administration of
organic compounds. In the trivalent state it can be bound mangafodipir. The results of these studies indicate that
986 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

single, clinical doses of mangafodipir are not likely to may undergo changes in oxidation state in the body.
cause persistent Mn accumulation in the brain. The oxidation state of the Mn ion in several enzymes
appears to be Mn(III), while most Mn taken in from
the environment is either Mn(II) or Mn(IV). Gibbons
6.2.1 Transport
et al. (1976) suggested that oxidation of Mn occurs in
Manganese homeostasis is vital for the optimal the body. It was observed that human ceruloplasmin
function of any organism, given the delicate relation- caused the oxidation of Mn(II) to Mn(III) in vitro and,
ship between its essentiality and toxicity. Manga- although the process has not been studied in vivo, it
nese (Mn2+) enters the portal circulation and binds to is a likely mechanism for Mn oxidation in the blood.
plasma α2-macroglobulin or albumin, its major carriers The same authors also noted that Mn oxidation led to
in blood. A small fraction of Mn in the 3+ oxidation a shift in Mn binding in vitro from α2-macroglobulin
state is bound to Tf. In the form of Mn3+, Mn is able to to Tf, and that in vivo clearance of Mn(II)-α2-
cross membranes via Tf receptor (TfR)-mediated endo- macroglobulin from cows was much more rapid than
cytosis (Aschner and Gannon, 1994). More than 99% of the clearance of Mn(III)-Tf. Another line of evidence
Mn is in the 2+ oxidation state. DMT-1, a nonspecific that Mn may undergo changes in oxidation state in
metal transporter, is the major transporter of Mn in this the body is based on measurements of Mn in tissues
oxidation state (Fitsanakis et al., 2007). and fluids using electron spin resonance (ESR), which
DMT-1 was previously referred to as NRAMP2 or detects the unpaired electrons in Mn(II), Mn(III), and
the divalent cation transporter (DCT1) because of its Mn(IV). When animals were injected with MnCl2, lev-
ability to transport cations, such as zinc (Zn2+), Mn2+, els of Mn increased in the bile and tissues, but only
cobalt (Co2+), cadmium (Cd2+), copper (Cu2+), nickel a small portion was in a form that gave an ESR sig-
(Ni2+), lead (Pb2+), and Fe2+ (Forbes and Gros, 2003; nal (Sakurai et al., 1985). This suggests that Mn(II) is
Gunshin et al., 1997; Knopfel et al., 2005). The DMT-1 converted to another oxidation state, probably Mn(III),
transporter family is highly conserved. DMT-1 ortho- although it is also possible that the formation of com-
logs are hydrophobic integral membrane proteins con- plexes between Mn(II) and biological molecules (e.g.
taining 11-12 transmembrane domains and they share bile salts, proteins, nucleotides) results in loss of the
a conserved “consensus transport sequence,” which is ESR signal without formal oxidation of the Mn ion.
involved in divalent metal ion translocation, including The extent of Mn redox reactions may be an impor-
that of Mn2+. The human gene spans an excess of 36 kb tant determinant of Mn retention and toxicity in the
and consists of 17 exons, encoding two proteins of 561 body. In a study by Komura and Sakamoto (1991), it
and 570 amino acids. The two alternatively spliced was shown that the form in which Mn was adminis-
mRNAs differ by a specific sequence that contains tered in the diet affected tissue levels of Mn in rats. In
or lacks an iron regulatory element (+IRE and −IRE, animals fed Mn acetate or MnCO3, levels of Mn were
respectively). The genes encode two transporters with significantly higher than in animals fed MnCl2 or
distinct C-termini (Fleming et al., 1998; Lee et al., 1998). MnO2. Differences in Mn concentrations in the blood
In addition to DMT-1, the choline transporter (Lock- and brain regions depending on the oxidation state of
man et al., 2001), voltage-gated and store-operated cal- Mn were also noted in another study (Roels et al., 1997).
cium channels (Lucaciu et al., 1997; Riccio et al., 2002), The MMT metabolites, hydroxymethylcyclopenta-
ionotropic glutamate receptor Ca2+ channels (Kannur- dienyl Mn tricarbonyl (CMT-CH2OH) and carboxycy-
patti et al., 2000), and the citrate transporter (Cross- clopentadienyl Mn tricarbonyl (CMT-COOH), appear
grove et al., 2003) have also been implicated in Mn2+ in urine following i.v. administration of MMT in male
transport. Additional evidence points to Mn trans- rats, as shown in a study by Hanzlik et al. (1980). These
port through the zinc transporters, ZIP8 and ZIP14 metabolites are also present in the bile, as indicated by
(Fujishiro et al., 2011; He et al., 2006), and ATP13A2, the fecal recovery of 3H from the ring structure in MMT
a P-type transmembrane ATPase protein (Gitler et al., following i.v. or intraperitoneal (i.p.) administration of
2009). Ferroportin-1 (Fpn) [also referred to as iron-reg- radiolabelled compounds to rats (Hanzlik et al., 1980).
ulated transporter 1 (IREG1) or metal tolerance protein Maneb and mancozeb are metabolized to several
1 (MTP1)] is the only transporter identified to date to compounds, including ethylenethiourea (ETU), ethyl-
outwardly transport Mn (Yin et al., 2010). eneurea, and ethylenediamine, in plants and animals.
ETU has been identified in the urine of occupation-
ally exposed sprayers of these fungicides (Kurttio and
6.3 Metabolism
Savolainen, 1990;, Kurttio et al., 1990).
Manganese is capable of existing in a number Mangafodipir, after infusion of the clinical dose of
of oxidation states, and limited data suggest that it 5 μmol/kg (or 0.25 mg/kg), is rapidly dephosphorylated
45 Manganese 987

to Mn dipyridoxyl monophosphate. The monophos- incompletely cleared from the body 24 h after admin-
phate then is fully dephosphorylated to Mn dipyridoxyl istration, with roughly 7-8% of the dose still being
ethylenediamine. This compound has been isolated in retained in the body after 1 week (Hustvedt et al., 1997).
blood from 18 min after the start of an infusion of man-
gafodipir until 40 min after the start (Toft et al., 1997). 6.5  Physiologically Based Pharmacokinetic
Model
6.4 Excretion A qualitative physiologically based pharmacoki-
Manganese homeostasis is primarily achieved by netic model for Mn toxicity in humans and animals
excretion. Absorbed Mn is almost totally excreted via was developed some years ago by Andersen et al.
the intestinal wall by several routes, and the main (1999). As several data gaps still exist concerning Mn
excretion route is via the bile. Manganese is removed pharmacokinetics, this model has been updated over
from the blood by the liver, where it conjugates with time, and it now includes models for adult, pregnant,
bile and is excreted into the intestine. After reaching lactating, and neonatal rats, nonhuman primates, and
the intestine, most of the element is excreted in the adult, pregnant, lactating, and neonatal humans. These
feces (Davis et al., 1993; Malecki et al., 1996). Liver models predict manganese tissue concentrations in
lysosomes play an important role in intrahepatic fetal, neonatal, pregnant, or aged individuals, as well
movement and affect the biliary excretion of absorbed as in individuals with liver disease or chronic man-
Mn (Suzuki and Wada, 1981). Some of the Mn in the ganese inhalation (Taylor et al., 2012; Dorman et al.,
intestine is reabsorbed through enterohepatic circula- 2012). When applied to monkeys, this analysis shows
tion. Small amounts of Mn can also be found in urine, that the dose-response relationship for the neurotoxic
sweat, and milk. In humans, the urinary excretion of effects of Mn is independent of exposure route, and
Mn is less than 1 μg/L. Since the body burden of an indicates the use of target tissue Mn concentration or
adult is about 10 mg Mn, only about 0.01% of the body cumulative target tissue Mn as the appropriate dose
burden is excreted daily via this route. metric for these comparisons (Schroeter et al., 2012).
Rats exposed to either MnCl2 or Mn3O4 by endo-
tracheal instillation excreted about 50% of the dose in 7  HEALTH EFFECTS
the feces within 3-7 days (Drown et al., 1986). Mon-
keys exposed to an aerosol of 54MnCl2 excreted most
7.1  Manganese Deficiency
of the Mn with a half-time of 0.2-0.36 days (Newland
et al., 1987). However, a portion of the compound was Manganese, like zinc and copper, is essential for nor-
retained in the lungs and brain. Clearance of this label mal prenatal and neonatal development. It is essential
was slower, occurring with half-times of 12-250 days. for humans and animals and plays a role in bone min-
Occupationally exposed male workers were eralization, protein and energy metabolism, metabolic
reported to have urine Mn levels significantly higher regulation, cellular protection from damaging free radi-
than those of unexposed persons (Roels et al., 1987b; cal species, and the formation of glycosaminoglycans
Alessio et al., 1989; Lucchini et al., 1995). Mergler et al. (Welder, 1994). Mitochondrial superoxide dismutase
(1994) did not report a significant difference in urinary (SOD), pyruvate carboxylase, and liver arginase are
Mn levels between exposed and control workers. The known Mn metalloenzymes (NAS/NRC, 1989; Welder,
differences in urinary excretion may be due to differ- 1994). Several enzyme systems: transferases, decarbox-
ences in the duration and extent of exposure. ylases, hydrolases, dehydrogenases, synthetases, and
Humans who ingested tracer levels of radioac- lyases, have been reported to interact with or depend
tive Mn, usually as MnCl2, excreted Mn with whole- on Mn for their catalytic or regulatory function (Welder,
body retention half-times of 13-37 days (Sandstrom 1994). Manganese has been shown to stimulate the syn-
et al., 1986; Davidsson et al., 1989). In a study by thesis of chondroitin sulfate, an important constituent of
Davis and Greger (1992), urinary excretion of Mn by the cartilage and connective tissue. There is limited evi-
men was 7.0 μmole/g creatinine and by women was dence that some disorders or diseases in humans such
9.3 μmole/day. Urinary excretion was not responsive as amyotrophic lateral sclerosis, acromegaly, catabolic
to oral intake of Mn. disease, and epilepsy may be associated with an imbal-
After exposure to the organic compound MMT, Mn ance in tissue levels of Mn (Carl et al., 1993). However,
excretion takes place to a large extent via urine and is no large-scale deficiency has been reported, although
of the same magnitude as GI excretion. Manganese Mn intakes of many people are believed to be less than
originating from clinical application of mangafodipir the estimated safe and adequate daily dietary intake
is primarily excreted in the feces via the bile and is (ESADDI). This may reflect, in part, the lack of adequate
988 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

methods to monitor nutritional status in regard to Mn. 7.3  Neurotoxic Effect


In a study by Friedman et al. (1987), five out of seven
CNS is the primary target of Mn toxicity. Although
men fed a Mn-deficient diet for 39 days exhibited der-
Mn is known to be a cellular toxicant that can impair
matitis (miliaria cristallina). The dermatitis cleared rap-
transport systems, enzyme activities, and receptor
idly when Mn was returned to the diet. Considering
functions, the principal manner in which Mn neuro-
the fact (supported by studies in humans) that Mn is an
toxicity occurs has not been yet clearly established
essential element, the U.S. Food and Nutrition Board of
(Aschner and Aschner, 1991).
the National Research Council established the ESADDI
levels as follows: 0.3-0.6 mg/day for infants from birth
7.3.1  Mode of Action
to 6 months; 0.6-1.0 mg/day for infants from 6 months
to 1 year; 1.0-1.5 mg/day for children from 1 to 3 years; Insight into which brain areas preferentially accu-
1.0-2.0 mg/day for children from 4 to 6 years, and from mulate Mn were obtained from autopsy samples from
7 to 10 years; and 2.0-5.0 mg/day for adolescents (> 11 individuals working in heavily contaminated environ-
years) and adults (NRC, 1989). However, the Food and ments (Yamada et al., 1986). These and other studies
Nutrition Board has cautioned that upper levels of these have shown that the corpus striatum and the extrapy-
provisional dietary intakes of ESADDIs should not be ramidal motor system are targeted by excessive expo-
exceeded on a routine basis because toxic levels may sure to Mn (Archibald and Tyree, 1987; Eriksson et al.,
be only a few times the usual intake levels (NRC, 1989). 1992b). Manganese concentrations vary across various
brain regions, attaining the highest level in the globus
pallidus (Prohaska, 1987), and concentrating predomi-
7.2  Acute Effects
nantly within the mitochondria of the basal ganglia
Manganese is less toxic than most metals. The (Morello et al., 2008; Liccione and Maines, 1988). Studies
average median lethal dose ranges from 400 to in nonhuman primates have shown by means of MRI
830 mg/Mn/kg (guinea pig, mouse, and rat) for oral the presence of Mn in the striatum, pituitary, and globus
administration of soluble Mn compounds and from pallidus, with preferential accumulation in the globus
38 to 64 mg Mn/kg (rat, mouse) for parenteral injec- pallidus, substantia nigra pars reticularis, and striatum
tions (Holbrook et al., 1975). Mn is regarded to be a (Newland et al., 1989). The specific area of injury par-
metal with relatively low toxicity; acute poisoning by allels the preferential accumulation of Mn within the
Mn in humans is very rare. Hobbesland et al. (1997) globus pallidus. Significant neuropathological changes
reported a significantly increased incidence in sudden such as cell loss and gliosis have been reported not only
death mortality for workers in Fe/Si-Mn plants during in the globus pallidus but also in the caudate, putamen,
their employment period [standardized mortality ratio and subthalamic nucleus (Yamada et al., 1986).
(SMR) 2.47]. However, these authors caution that the The substantia nigra may be affected by Mn, but
association between increased death rate and Mn expo- generally to a lesser extent than the globus pallidus
sure is speculative, and that the incidence in sudden (Katsuragi et al., 1996; Newland et al., 1989; Yamada
death could also be caused by common furnace work et al., 1986). Positron emission tomography (PET)
conditions (e.g. heat, stress, noise, carbon monoxide). studies have shown that Mn does not alter the integ-
There is only one study (Kawamura, 1941) in which rity of nigrostriatal dopaminergic neurons (Shinotoh
death in humans may have been associated with inges- et al., 1995). In general, it appears that the substantia
tion of Mn. In this report, death from “emaciation” nigra pars compacta (SNpc), the primary area affected
occurred in two adults who ingested drinking water con- in Parkinson disease (PD), is largely intact after Mn
taminated with high levels of Mn (14 mg/L). Although exposure (Yamada et al., 1986). However, insult to
many of the symptoms in the group of six Japanese fam- the globus pallidus by Mn may affect SNpc through
ilies (about 25 persons) could be connected with expo- downstream pathways, consistent with the occurrence
sure to Mn, there is considerable doubt that all of the of parkinsonian symptoms (Verity, 1999).
features of this outbreak (particularly the deaths) were There is limited evidence that dopamine levels in
due to Mn alone (ATSDR, 2012). Namely, symptoms the caudate nucleus and the putamen are decreased in
appeared to have developed very quickly: the course affected patients. Dopamine is one of the most com-
of the disease was very rapid, in one case progressing mon catecholamines and it functions to control loco-
from initial symptoms to death in 3 days; and all survi- motion, emotions, and neuroendocrine secretion. To
vors recovered from the symptoms even before the Mn date, most emphasis on the neurotoxic effects of Mn
content of the well had decreased significantly after the has been directed at its ability to alter the dopamine
removal of batteries (from which Mn had leached to the transporter (DAT) or dopamine receptor (DAR). Sev-
drinking water) buried near the well. eral studies have shown that postnatal exposure to Mn
45 Manganese 989

causes a decrease in presynaptic dopaminergic function et al., 2000). Furthermore, given the important role of
through reducing expression of the DAT protein and glutamate in mediating neurotransmission, this amino
uptake of dopamine in the striatum, as well as decreas- acid is highly sensitive to changes in energy supply
ing long-term dopamine efflux (Huang et al., 2003; (see below). Overactivation of ionotropic glutamate
McDougall et al., 2008). Additional evidence suggests receptors (GluRs) by Mn leads to increased energy
that dopaminergic neurotransmission is attenuated by consumption, which may lead to accelerated depletion
Mn, concomitant with depletion of striatal dopamine of ATP levels and a generalized failure in maintaining
(Calne et al., 1994; Pal et al., 1999; Reichel et al., 2006; optimal cellular energy stores.
Chen et al., 2008). It has also been shown that Mn can Several studies also point to an effect of Mn on
affect the dopamine D1 receptors in the striatum of γ-aminobutyric acid (GABA). GABA content (Bonilla,
monkeys (Eriksson et al., 1992a), as well as the density 1978; Fitsanakis et al., 2006; Gwiazda et al., 2007) and its
of D2 receptors in developing rats (Seth and Chandra, release have been shown to increase in response to Mn
1984). The valence of Mn may influence its toxicity as (Prestifilippo et al., 2008). It has also been demonstrated
to dopamine. In vitro, Mn3+ appears more toxic than that striatal GABA-containing neurons show height-
Mn2+, possibly due to higher oxidative reactivity and a ened sensitivity to Mn compared to mesencephalic
closer radius resemblance to iron. In biological media, dopaminergic neurons (Defazio et al., 1996). Other stud-
Mn in the 3+ oxidation state reacts with dopamine ies showed inhibition of GABA uptake in response to
more potently than it does in the 2+ state, catalyzing Mn treatment (Anderson et al., 2007), with diminished
its auto-oxidation to reactive quinones that generate pallidal GABA concentration (Struve et al., 2007), while
ROS, resulting in oxidative stress, lipid peroxidation, no changes in GABA levels were reported by Garcia
and cellular damage (Lloyd, 1995; Shen and Dryhurst, and colleagues (2007). These discrepancies may reflect
1998; Ahmadi et al., 2008; Stokes et al., 1999). The auto- a temporal factor that contributes to these effects. They
oxidation of dopamine may lead to the formation of may also reflect differences in treatment regime (con-
highly reactive semiquinone and aminochrome inter- centration and time of dose) as well as species varia-
mediates (Baranyi et al., 2006; Shen and Dryhurst, 1998; tions; however, in general they are consistent with the
Lloyd, 1995; Graumann et al., 2002; Florence and Stau- vulnerability of GABA-containing regions to Mn.
ber, 1989; Oikawa et al., 2006). The loss of dopamine While the cholinergic system does not appear to
in the brain and the concomitant neuronal cell dam- be a primary target of Mn toxicity, it may affect pre-
age might be reflected as an increase in motor activity synaptic choline uptake (Lockman et al., 2001; Eriks-
(Bonilla, 1984; Nachtman et al., 1986). Calabresi et al. son et al., 1984; Lai et al., 1984), acetylcholine (ACh)
(2001) also reported that Mn-treated rats exhibited a transferase activity (Lai et al., 1984), the release of
complex behavior syndrome, with increased activity ACh (Kita et al., 1981), postsynaptic ACh binding to
in the absence of significant striatal neuronal loss and receptors, and ACh clearance by acetylcholinesterase
gliosis. Studies in Mn-exposed nonhuman primates (Lai et al., 1992). Moreover, Mn influences ACh bind-
have demonstrated a lack of nigrostriatal dopaminer- ing and transport proteins in astrocytes (Finkelstein
gic degeneration in the striatum; however, these ani- et al., 2007). Changes in the levels of this neurotrans-
mals do express dopamine neuron dysfunction: there mitter are consistent with the early psychotic phase
is marked inhibition of in vivo dopamine release in the of manganism, referred to as “Mn madness” as well
striatum as measured by PET (Guilarte et al., 2008), a as cognitive deficits in memory and intellectual decay
finding that has been confirmed in rodent models of (Feldman and Ratner, 1999; Iregren, 1999; Tanner, 1992;
Mn exposure (Peneder et al., 2011). Wennberg et al., 1991), and deficits in audioverbal
In addition to the dopaminergic system, other neu- short-term memory and hand-eye coordination (Roels
rotransmitter systems have been implicated in the et al., 1987b; Mergler et al., 1994).
neurotoxicity of Mn. Glutamate uptake by astrocytes is It is also thought that ACh activity may impact
significantly reduced by Mn, thus increasing the extra- the dopaminergic system (Eells and Brown, 2009).
cellular concentration of glutamate and its excitotoxic- Increased ACh levels in the cortex, striatum, and hippo-
ity. This effect is probably mediated by the propensity campus have been reported (Fosbraey et al., 1990). Since
of Mn to decrease the expression of genes encoding the these areas also contain dopaminergic neurons prone to
astrocytic glutamate transporters, such as excitatory degenerate upon Mn exposure, and since dopamine is
amino acid transporters 1 and 2 (SLC1A3/GLAST known to affect ACh release by the septohippocampal
and SLC1A2/GLT1) (Erikson and Aschner, 2002, 2003; and striatal cholinergic neurons (Nilsson et al., 1992;
Erikson et al., 2002b). Manganese may also increase Robertson et al., 1992; Sato et al., 1994), this relationship
the sensitivity of postsynaptic receptors to glutamate, between dopamine and ACh may explain the symptom-
thus activating Mn-affected pallidal neurons (Spadoni atology and clinical picture inherent in Mn exposure.
990 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

Mitochondria are the target organelles of Mn. Man- and in ferroalloy industry was the most common cause
ganese has a greater affinity than Ca2+ for the calcium of Mn clinical intoxication from Chile, Cuba, Germany,
uniporter, thus favoring its influx. Furthermore, once India, Mexico, Morocco, Romania, Sinai, Spain, and
within mitochondria, Ca2+ has greater affinity for the the former U.S.S.R. (WHO, 1981).
efflux transport. Therefore, Mn is essentially trapped The clinical features of classic manganism include
within the mitochondria, readily affecting its function psychiatric and neurological manifestations related
(Gavin et al., 1992). Manganese triggers mitochondrial to the extrapyramidal system. The disease typically
dysfunction through several mechanisms, predomi- begins with feeling of weakness and lethargy. As the
nantly via oxidative stress. Mn can impair mitochon- disease progresses, a number of other neurological
drial function by inhibition of mitochondrial electron signs become manifest. Although not all individuals
transfer chain, affecting the activity of respiratory develop identical signs, the most common are a slow
chain enzymes, inhibiting complexes I-IV, and inter- and clumsy gait, speech disturbances, a mask-like
fering with oxidative phosphorylation (Zhang et al., face, and tremors. The speech disorder may progress
2004). In addition, Mn can bind to a specific magne- to muteness. Patients develop a fixed facial expres-
sium (Mg2+)-binding site on the F1-ATPase, thereby sion. Increasing clumsiness and an inability to make
slowing its activity (Smith et al., 1985) and effectively certain movements have also been observed. In the
reducing ATP production. This leads to increased leak- third stage, the disease is fully developed. Difficulties
age of electrons and increased superoxide (O2•−) pro- in walking caused by muscular hypertonia, muscle
duction, with eventual collapse of cellular function, rigidity, and tremor in the upper limbs, as well as diffi-
which may be followed by cell death (Scholte, 1988). culties in writing, are among the symptoms and signs.
In addition, Mn can alter calcium homeostasis in In addition, a syndrome of psychological and psychic
mitochondria by inhibiting its efflux and increasing disturbances (aggressiveness, hallucination, psychosis)
its levels in the organelle (Gavin et al., 1990; Spadoni frequently emerges. Although neurological symptoms
et al., 2000). Elevated calcium levels per se may trigger may improve in some cases (Ky et al., 1992), clinical
excessive ROS production with subsequent opening of progression is irreversible, persisting after exposure
the mitochondrial permeability transition pore (MTP), ceases (Huang et al., 1998). Individual susceptibility is
thereby increasing the permeability to protons, ions, thought to play a role in the development of mangan-
and solutes (Zoratti and Szabo, 1995). These effects ism, as it occurs after only a few months of exposure,
culminate in the loss of the inner membrane potential whereas in many workers with more than 20 years of
(Δψm), mitochondrial swelling, and finally impaired exposure, no adverse neurological effects have been
oxidative phosphorylation and ATP synthesis (Gunter observed (WHO, 1981).
and Pfeiffer, 1990). Differences between the classical features of man-
Manganese has also been shown to trigger apop- ganism and PD have been described. Barbeau (1984)
tosis. Manganese-induced MTP opening leads to the reported that dystonia, hypokinesia, and intentional
activation of Bcl-2 family proteins, especially Bax/Bak, tremor presented in patients suffering from mangan-
culminating in the release of cytochrome c and activa- ism more frequently than in parkinsonian patients.
tion via the extracellular signal-regulated kinase (ERK) Psychiatric disturbances early in the disease (although
pathway of the cysteine protease caspase-3, the apop- not always), a cock-walk, a propensity to fall backward
totic executioner protein. It has been shown that Mn when displaced, less frequent resting tremor, more fre-
is a potent inducer of ERK and caspase-3 activation quent dystonia, and failure to respond to dopamino-
(Malecki, 2001), consistent with observations of DNA mimetics, are other characteristics that distinguished
strand breakage [Malecki, 2001]. manganism from PD (Calne et al., 1994). In PD patients,
pathological lesions are found in the substantia nigra
and other pigmented areas of the brain (Barbeau, 1984).
7.3.2  Health Impairment
Parkinsonism is due to the selective loss of subcortical
As early as 1837, John Couper reported five cases of neurons, whose cell bodies lie in the substantia nigra
“manganism” among men employed in grinding Mn and whose axons terminate in the basal ganglia, which
dioxide in the manufacture of chlorine for bleaching includes the caudate nucleus, the putamen, the globus
powder (Couper, 1837). The clinical picture was similar pallidus, and other structures. These nigral neurons
to paralysis agitans and was published only 20 years use dopamine as their neurotransmitter. Manganese
after Sir James Parkinson’s description of the “shaking appears to affect pathways that are postsynaptic to the
palsy.” According to the EPA (1993), 7550 cases of man- nigrostriatal system, mostly the globus pallidus (Chu
ganism had been recorded since the first report in 1837. et al., 1995). MRI of the brain reveals the accumulation
Exposure to Mn in mines or in ore processing plants of Mn in cases of manganism, but few or no changes
45 Manganese 991

in subjects with PD. 18Fluorodopa (F-dopa) PET scans (CI) 1.41-1.76] compared to the other counties in the
were generally normal in cases of manganism but same province (crude prevalence 246/100.000). This
abnormal in subjects with PD (Calne et al., 1994). observation may indicate the interaction of prolonged
However, this paradigmatic description of differ- environmental exposure to Mn with genetic factors,
ences and similarities has evolved until now there is potentially relevant in this mountain population (Luc-
a less clear distinction made between the two clinical chini et al., 2007). A study by Finkelstein and Jerrett
manifestations. Preventive measures have virtually (2007) of the relationship between PD and markers
eliminated the possibility of uncontrolled occupational of traffic-derived and environmental Mn air pollu-
exposure to high levels of Mn. Therefore, outbreaks of tion in two Canadian cities has shown no association
classical manganism are no longer reported. Prolonged in Toronto (only traffic-associated air-Mn), whereas
and lifetime exposures to lower levels have become in Hamilton (both traffic and industry-associated air-
the more common situation (Lucchini and Zimmer- Mn) the odds for a physician’s diagnosis of PD was
man, 2009), related to a parallel increase of Mn levels 1.034 (95%CI 1.00-1.07) per 10 ng/m3 increase of Mn in
in the environment (Herndon et al., 2011). In this con- TSP. The prevalence curves suggested that exposure to
text, damage induced by Mn can also extend into the ambient Mn advances the age of PD diagnosis, which
dopaminergic pathways that are typically damaged in is consistent with the theory that exposure to Mn adds
PD (Baek et al., 2004; Kim et al., 2002; Lucchini et al., to the natural loss of neurons attributable to the aging
2009). Typical signs of Mn deposition at MRI scan, with process.
elevated B-Mn and abnormal F-dopa PET scan, have An increased frequency of parkinsonian distur-
been shown in welders (Kim et al., 1999a). A patient bances has also been shown in case-control studies on
with parkinsonism associated with elevated serum large groups of welders (Racette et al., 2005b). The esti-
Mn from hepatic dysfunction demonstrated relatively mated prevalence of parkinsonism among active male
symmetrically and severely reduced F-dopa levels in welders aged 40-69 years was 977-1336 cases/100,000
the posterior putamen. MRI revealed selective abnor- population, and was significantly higher compared to
mality within the internal segment of the pallidum age-standardized data for the general population (prev-
(Racette et al., 2005a). Reduced uptake of F-dopa is alence ratio 10.19; 95% CI 4.43-23.43). The parkinsonian
also shown in end-stage liver disease (Criswell et al., features in welders seem not to be different from those
2011) and in welders (Criswell et al., 2012a) in rela- of PD, except for a younger age of onset (Racette et al.,
tion to Mn exposure. Elevated T1-weighted intensity 2001). Studies in Korea (Park et al., 2006) and Sweden
indices and increased apparent diffusion coefficients at (Fored et al., 2006) did not observe an increased preva-
MRI are shown throughout the basal ganglia in weld- lence of disturbances in welder cohorts. An increased
ers (Criswell et al., 2012b). risk of parkinsonism has been further confirmed in a
Manganese can be considered an environmen- large cohort of 811 shipyard and fabrication welders
tal trigger of PD or parkinsonian disorders that can and associated with weighted welding exposure-years
accelerate the onset of neurodegenerative damage (Racette et al., 2012). Parkinsonism (rigidity, bradyki-
(Martin, 2006) by interacting with genetic factors (Le nesia, rest tremor, and postural instability) is present
Couteur et al., 2002; Zatta et al., 2003). The relationship in 15% of Mn-exposed workers in welding indus-
between these two manifestations has triggered fur- tries; these parkinsonian signs also overlap consider-
ther research, also in view of the close interconnection ably with the clinical findings seen in early stages of
between the target area of Mn toxicity (represented by PD, although molecular imaging suggests that Mn-
the globus pallidus) and the substantia nigra, where exposed workers have dopaminergic dysfunction in a
the degeneration of dopaminergic neurons takes place pattern that differs from PD (Racette et al., 2013).
(Weiss, 2006). Studies to estimate the impact of occupational expo-
Elevated environmental Mn exposure may affect sure to Mn on neurological health have employed
nonoccupational populations living in the vicini- sensitive tests designed to detect early signs of neuro-
ties of elevated Mn emissions. Studies have shown psychological and neuromotor deficit (Iregren, 1999).
an increased risk of PD or parkinsonian disturbances Dose-response relationships were shown when expo-
in older adults living in areas with elevated envi- sure was expressed with cumulative exposure indices
ronmental Mn (Lucchini et al., 2007; Finkelstein and (Roels et al., 1987a, 1992; Lucchini et al., 1995, 1999).
Jerrett, 2007). The frequency of parkinsonian distur- Average Mn levels in the air reported to lead to early
bances in the surroundings of ferroalloy plants in an signs of nervous system toxicity after inhalation expo-
Italian prealps valley (crude prevalence 358/100,000 sure range from 0.027 to 1 mg Mn/m3 (Chia et al., 1993,
population; standardized for age and sex 438) was sig- 1995; Iregren, 1990; Mergler et al., 1994; Lucchini et al.,
nificantly higher [SMR 1.58; 95% confidence interval 1995, 1997, 1999; Roels et al., 1987a, 1992; Wennberg
992 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

et al., 1991). Overt manganism was observed at expo- increased tremor, reduced response speed, enhanced
sure levels ranging from 2 to 22 mg Mn/m3 (Šarić olfactory sense, possible memory and intellectual
et al., 1977). According to World Health Organization deficit, and mood changes. Several studies suggest the
(WHO, 1981), clinical manifestation of Mn intoxication existence of dose-effect relationships, such as between
can occur at 1 mg/m3 in susceptible individuals. Mn exposure (cumulative air-Mn exposure index or
Roels et al. (1987b) detected early preclinical neuro- B-Mn) and neuromotor or neuropsychological (cog-
logical effects in > 25% of a group of workers exposed nitive) test results, for example in 43 confined space
to 0.97 mg Mn/m3 (median concentration in total dust) welders exposed to Mn welding fume for 1.5 years
for 7.1 years on average. Similarly, Iregren (1990) used on average (Bowler et al., 2007). The mean TWA of
neurobehavioral tests (simple reaction time, digit span, air-Mn ranged from 0.11-0.46 mg/m3 (overall mean
finger tapping, verbal ability, hand dexterity, and finger 0.21 mg/m3; 55% of the personal air samples >0.20 mg/
dexterity tests from Swedish performance evaluation m3). A follow-up study in this welder cohort (n = 26)
system) to study adverse effects in 30 male workers after 3.5 years of reduced or cessation of Mn exposure,
from two different Mn foundries exposed to an esti- revealed that only cognitive function improved sig-
mated median concentration of 0.14 mg Mn (in total nificantly, while olfactory, extrapyramidal, and mood
dust)/m3 as MnO2 for 1-35 years. The exposed workers disturbances remained the same or were exacerbated.
had below average scores on a number of the tests, such This suggests differential intrinsic vulnerabilities of
as reaction time and finger tapping, when compared brain loci involved in Mn exposure (Bowler et al., 2011).
to matched controls. Dose-dependent (cumulative Mn A meta-analysis of performance effects resulting
exposure) worsening of eye-hand coordination, hand from occupational exposure to Mn has been conducted
steadiness, and visual reaction time were reported in on 13 epidemiological studies published between 1987
dry battery workers exposed on average to about 1 mg and 2008 (Meyer-Baron et al., 2009). Mean concentra-
Mn/m3 in total dust for about 5 years (Roels et al., tions of inhalable Mn ranged from 0.05 to 1.59 mg/m3,
1992). In the subsequent 8-year follow-up study by and mean concentrations of Mn in whole blood ranged
Roels et al. (1999), only the low-exposure group showed from 8.1 to 48.4 μg/L. Nineteen neuropsychological
normalization of performance in the eye-hand coordi- performance variables were analyzed and, apart from
nation test following a decrease in the average annual two outcomes, the overall effects displayed a negative
Mn level in total dust from 0.471 to 0.158  mg/m3. impact of Mn on performance. Significant overall defi-
The medium- and high-exposure groups showed a cits were obtained for six test variables; their effect size
persistent performance deficit after this period, during ranged from d = −0.23 to −0.36. Four of the variables
which the average annual air-Mn levels dropped from measured motor speed and two of them speed of infor-
0.818 to 0.241 mg/m3 and from 2.941 to 0.744 mg/m3, mation processing. The analysis of exposure-effect
respectively (Roels et al., 1999, 2012). None of the relationships showed that larger effect sizes were more
exposure groups showed a significant improvement in consistently associated with higher concentrations of
hand stability and simple visual reaction time. In the inhalable Mn than with Mn in blood. Confounding
study by Mergler et al. (1994), the Mn-exposed work- from individual-level and study-level covariates was
ers had similar deficits and also differed significantly shown by analyses of variance, but could be reduced
from the controls in cognitive flexibility and emo- by linear regressions and z-normalization using data
tional state. They also exhibited significantly greater from the respective control groups (Meyer-Baron et al.,
levels of tension, fatigue, and confusion, as well as a 2011). Results from the meta-analysis prompted a
significantly lower olfactory threshold than controls. pool analysis of data from eight studies. Four cogni-
Workplace levels of Mn in this study were measured tive and motor performance variables each indicated
at 0.014-11.48 mg/m3, while Mn levels in the respirable significantly lower performances of exposed individu-
fraction were 0.001-1.273 mg/m3, and mean duration als when confounding was considered; slowed motor
of exposure was 16.7 years. Epidemiological studies performances and deficits in attention and short-term
by Lucchini et al. (1995, 1997, 1999) using neuropsy- memory were found (Meyer-Baron et al., 2013).
chological tests have shown similar findings on neu- There is evidence to suggest that Mn neurotoxicity
robehavioral and motor function. These studies have can result also from prolonged exposure to much lower
also shown dose-effect relationships with cumulative Mn exposure levels, such as those occurring in the gen-
exposure indices. Mergler and Baldwin (1997) pointed eral population. This has been studied in residents liv-
out that although outcomes from individual studies ing in the proximity of industries emitting Mn into the
using neurophysiologic and neurobehavioral tests in environment. In 1999, there was a dose-effect associa-
the absence of clinical manifestation can vary, they col- tion between Mn in blood and several neurobehavioral
lectively show a pattern of slowing motor functions, tests examining motor and cognitive functions and
45 Manganese 993

mood in residents in the vicinity of a former ferro-Mn children whose drinking water contained high concen-
alloy plant in South Quebec. These effects were more trations of Mn. A study of the association between Mn
pronounced in individuals over 50-year old and in men exposure through drinking water and infant mortality
(Mergler et al., 1999). Santos-Burgoa et al. (2001) pub- (Hafeman et al., 2007) showed that infants exposed to
lished a cross-sectional study into the health effects of water Mn concentrations greater than or equal to the
Mn in a group of adult subjects from a mining district WHO (2004b) standard of 0.4 mg Mn/L had an ele-
in Central Mexico with different levels of airborne Mn. vated mortality risk during the first year of life com-
Using a multivariate logistic regression analysis, the pared with unexposed infants.
authors identified a significant relationship between A study conducted in Bangladesh showed that Mn
deficient cognitive performance (Mini-Mental score of may affect intellectual function, resulting in lower
less than 17) and Mn blood levels. The same group has intelligence quotient (IQ) (Wasserman et al., 2006).
confirmed that impaired motor functions in residents This study examined a cross-section of 142 10-year old
in the vicinity of Mn mines is associated with the air- children whose well water supply was contaminated
borne concentration of Mn, which had an average GM with a mean concentration of 793 μg Mn/L. The data
of 0.13 μg/m3 as (Rodriguez-Agudelo et al., 2006). Sub- indicated that Mn exposure was significantly associ-
clinical findings on motor functions, measured using ated with reduced full-scale, performance, and verbal
the Unified Parkinson’s Disease Rating Scale and raw scores on the Wechsler Intelligence Scale for Chil-
postural sway, were observed among a group of 100 dren in a dose-response fashion. The same population
residents from Marietta (Ohio, USA) living an aver- also showed an association between an impairment of
age of about 7 km from a ferro-Mn alloy smelter (Kim children’s mathematical academic achievement and
et al., 2011b). The modeled residential air-Mn estimate Mn in drinking water (Khan et al., 2012).
ranged from 0.04 to 0.96 μg/m3 (average 0.18 μg/m3). A study conducted to explore the potential associa-
Although no substantial differences in neuropsycho- tions between hair metal levels and the neuropsycho-
logical test scores were found compared to a group of logical function and behavior of school-aged children,
matched residents of an Ohio control town, the results showed that children’s general intelligence scores,
suggest the existence of some association of anxiety particularly verbal IQ scores, were inversely related
states and neuropsychological function with environ- to Mn and arsenic levels in hair. Significant interaction
mental air-Mn exposure at the Marietta site. It remains between these two metals was observed. The mean hair
an open question whether these associations are due metal levels were 471.5 parts per billion (ppb or μg/kg)
to direct neurotoxic effects of air-Mn or to the Marietta for Mn and 17.8 ppb for arsenic (Wright et al., 2006).
residents’ concern about the potential health effects Additionally, a study conducted in Quebec, Canada,
of air pollution (Bowler et al., 2012; Roels et al., 2012). examined the relationship between chronic exposure to
Impairment of motor coordination and odor identifica- levels of water Mn and hyperactive behavior in children
tion were observed among elderly residing in the vicin- (Bouchard et al., 2007). The same group observed intel-
ity of historical ferro-Mn plant sites in Valcamonica, lectual impairment (i.e. lowered IQ score) in 362 chil-
Italy (Zoni et al., 2012). Polymorphisms rs4920608 and dren exposed to a median Mn concentration in home
rs2871776 of the PD-related gene ATP13A2/PARK9 tap water of 34 μg Mn/L (range 1-2700 μg Mn/L). Man-
(encoding probable cation-transporting ATPase 13A2) ganese in water was correlated with Mn in hair and both
significantly modified the effects of Mn exposure were negatively associated with the IQ score (Bouchard
on impaired motor coordination in these subjects et al., 2011). The hair Mn concentration correlated with
(Rentschler et al., 2012). hyperactive and oppositional behaviors, with the high-
Manganese can also cause neurotoxic effects in exposure group having significantly greater levels of
young children, who may represent a more sensitive Mn in hair and a stronger association with hyperactive
subgroup of the general population because they show behaviors. Likewise, inhaled Mn was negatively associ-
increased absorption of Mn compared to adults (Dorner ated with intellectual function in school-aged children
et al., 1989). Except for recent reports of impaired motor living near a Mn mining and processing facility (Riojas-
skills and odor identification (Lucchini et al., 2012; Rodriguez et al., 2010). Recent findings show that mice
Zoni et al., 2012), health effects associated with ele- exposed to Mn during juvenile development experi-
vated Mn exposure in children are impaired cognitive enced greater neuroinflammatory injury and behav-
function, with decrements in memory, verbal learn- ioral dysfunction upon exposure to Mn later in adult
ing, and intelligence (Wasserman et al., 2006; Wright life than mice without juvenile Mn exposure (Moreno
et al., 2006; Bouchard et al., 2011; Riojas-Rodriguez et al., 2009). Hence, a background of Mn exposure, par-
et al., 2010; Menezes-Filho et al., 2011, 2013; Khan et al., ticularly during critical developmental years, could ren-
2012). Some of these studies reported neurotoxicity in der individuals more susceptible to neurotoxic insults
994 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

later in life that predispose to neurological disease. The early Mn intoxication because it may aggravate manic
literature on pediatric exposure indicates that cognitive symptoms due to the increase in brain dopamine.
functions also form an important target of Mn toxicity Patients affected by classic manganism typically do
(Zoni et al., 2007; Roels et al., 2012). The cerebral cortex not respond to l-dopa treatment (Huang et al., 1989;
is probably a critical target area, in addition to the basal Calne et al., 1994; Chu et al., 1995; Koller et al., 2004),
ganglia. Working memory seems to be affected by Mn indicating that they likely suffer degeneration of the
toxicity in the caudate nucleus in the striatum, frontal receptors and neurons that normally respond to this
cortex, and parietal cortex, as shown in nonhuman neurochemical (Chu et al., 1995). New data show, con-
­primates (Guilarte, 2013). versely, that l-dopa treatment reverses Mn-induced
Children who suffer from cholestatic liver disease motor alterations in mice after inhalation of 0.04 M
or who are under total parenteral nutrition for GI dis- Mn chloride and Mn acetate for 1 h twice a week for 5
orders are at risk of Mn overload (Devenyi et al., 1994; months (Ordoñez-Librado et al., 2010).
Fell et al., 1996; Ono et al., 1995). Impaired biliary Chelating agent therapy has also been tried, but
excretion entailed signs of manganism (Fell et al., 1996; mainly without success. Nevertheless, recent reports
Kafritsa et al., 1998; Ono et al., 1995; Nagatomo et al., indicate that chelation treatment may improve the
1999). High levels of brain Mn have been shown in clinical features in overexposed welders, when
patients with liver dysfunction, referred to as hepatic administered in the early phases of the clinical onset
encephalopathy (Butterworth et al., 1995; Butterworth, (Discalzi et al., 2000). In a follow-up of seven patients
2000, 2010; Zerón et al., 2011). affected by Mn-induced parkinsonism after treatment
Mutations in the SLC30A10 gene (encoding a man- with CaNa2 ethylenediaminetetraacetic acid (CaNa2
ganese transporter) were discovered to cause a syn- EDTA; edetate disodium calcium), good clinical, bio-
drome of hepatic cirrhosis, early onset generalized logical, and neurological results were obtained by a
dystonia, polycythemia, and hypermanganesemia combination of timely removal from exposure and
(Stamelou et al., 2012). This rare disorder, together chelating therapy (Herrero Hernandez et al., 2006).
with Wilson disease, is the only potentially treatable Sodium p-aminosalicylic acid has been observed to be
inherited metal storage disease to date, and it can be also effective in treating Mn-induced clinical manifes-
fatal as a result of complications of cirrhosis. tations in severe cases (Jiang et al., 2006). Chelation
Manganese poisoning may also occur upon abuse therapy with edetate disodium calcium combined
of illicit drug such as methcathinone (or ephedrine). with iron supplementation is the treatment of choice
Methcathinone is a homemade drug derived from the to lower B-Mn and improve clinical symptoms asso-
oxidation of pseudoephedrine with potassium per- ciated with the newly recognized metabolic disorder
manganate (Sikk et al., 2011). The source of the high characterized by hypermanganesemia, liver cirrhosis,
level of Mn in these preparations is residual potas- extrapyramidal motor manifestations, and polycythe-
sium permanganate, which is used in the synthesis mia (Tuschl et al., 2012).
of these drugs (Sikk et al., 2011). Critically premature
neonates are commonly fed total parenteral nutrition
supplemented with variable amounts of Mn. Hepatic 7.4  Effect on the Lungs
dysfunction and cholestasis are known risk factors of
7.4.1  Mode of Action
increased brain Mn accumulation (Butterworth, 2010;
Butterworth, et al., 1995). Acute or intermediate exposure to excess Mn affects
the respiratory system. Inhalation exposure to high
concentrations of Mn dusts (Mn dioxide and Mn tetrox-
7.3.3  Treatment of Manganism
ide) can cause an inflammatory response in the lung
The similarity of manganism to PD prompted Mena (chemical pneumonitis). However, exposure to Mn in
et al. (1970) to administer the amino acids l-3,4-dihy- an insoluble form, such as MnO2, may also be harmful
droxyphenylamine (levodopa, l-dopa) and 5-hydroxy- for lung also at low exposure levels. As quoted in the
tryptophan—precursors of dopamine and serotonin, WHO Regional Office for Europe document (2000), an
respectively—for the treatment of chronic Mn poison- increase in pneumonia incidence has been registered
ing. Barbeau (1984) also reported that oral l-dopa can in rats exposed to 43-139 μg Mn/m3 as MnO2 (mean
temporarily improve the symptoms of Mn-induced mass median aerodynamic diameter 0.76 μg; mean
neurotoxicity. In an experimental study on rats, Shukla standard geometric deviation 2.28) for 2 weeks (Shio-
and Chandra (1981) found that l-dopa treatment mark- tsuka, 1984), and pulmonary emphysema in monkeys
edly increases striatal dopamine level. Therefore, they exposed to 0.7-3.0 mg Mn/m3 as MnO2 (80% < 1 μm)
advised that l-dopa therapy should not be used in for 10 months (Suzuki et al., 1978).
45 Manganese 995

In order to obtain information on the histological An increased incidence of pneumonia, as well as bron-
characterization of pulmonary responses induced by chitis, was found in workers exposed to Mn concentra-
Mn, Dorman et al. (2005) performed an experimental tions of 0.39-16.35 mg/m3 in a factory producing Mn
study on rhesus monkeys exposed by inhalation to Mn alloys (Šarić et al., 1977). In this study, smoking hab-
sulfate at 0.06, 0.3, or 1.5 mg Mn for 65 exposure days. its were taken into account. Roels et al. (1987b) found
Animals exposed to Mn sulfate at ≥ 0.3 mg Mn/m3 had in workers of a Mn oxide and salt production plant a
increased lung Mn concentration. Exposure to Mn sul- slight but significant decrease in spirometric values
fate at 1.5 mg Mn/m3 for ≥ 15 exposure days resulted in [forced vital capacity, forced expiratory volume in 1
increased lung Mn concentration, mild subacute bron- second (FEV1)], whereas at a similar average air-Mn
chiolitis, alveolar duct inflammation, and proliferation level of about 1 mg/m3 no spirometric changes were
of bronchus-associated lymphoid tissue. Bronchiolitis found in workers of a dry alkaline battery exposed
and alveolar duct inflammatory changes were absent only to MnO2 (Roels et al., 1992). In both studies, smok-
45 days postexposure, suggesting that these lesions ing habits were accounted for and no interaction was
are reversible upon cessation of subchronic high-dose found between smoking and Mn exposure regarding
Mn exposure. Subchronic exposure at concentration the spirometric test outcome. Based on the spirometric
≤ 0.3 mg Mn/m3, close to the current 8-h occupational performance of the control groups in both studies, it is
TLVs established for inhaled Mn, was not associated likely that the workers from the dry battery plant had
with pulmonary pathology. intrinsically a much better respiratory function than
A toxicologic in vitro study (Jang, 2009) on prosta- those of the Mn oxide and salt production plant.
glandin G/H synthase 2/cyclooxygenase-2 (COX-2), Studies on populations living in the vicinity of Mn-
which is involved in the biosynthesis of inflammatory emitting plants indicated that Mn exposure might affect
prostaglandins, investigated the effect of Mn chloride the respiratory system. Effects at lower levels of air-
on COX-2 expression in A549 human lung epithe- borne Mn have been claimed (Šarić et al., 1978) to occur
lial cells. The data obtained showed that Mn induces in a population living near a ferro-Mn plant (Šibenik,
COX-2 expression by transcriptional upregulation in Croatia). Over a period of 4 years, acute bronchitis
A549 cells and that the induction is mediated through and peribronchitis were consistently more common
multiple cellular events. The findings may provide a among people living near the plant. A less consistent
molecular basis for Mn-associated airway inflamma- pattern could be seen for pneumonia. Average annual
tion via COX-2 upregulation. Mn concentrations in two zones of the town area
Laboratory animal studies have also shown that near the plant were 0.236-0.39 and 0.164-0.243 μg/m3,
inhaled Mn may increase susceptibility to infec- whereas in a third zone at a distance of 4-5 km from
tious agents such as Streptococcus pyogenes in mice the plant the annual means for 4 years varied from
(Adkins et al., 1980), Enterobacter cloacae in guinea pigs 0.042 to 0.099 μg/m3. The highest weekly means for the
(Bergström, 1977), and Klebsiella pneumoniae in mice zones were 1.24, 1.08, and 0.27 μg/m3, respectively. In
(Maigetter et al., 1976). There is sufficient evidence addition, in a study carried out in school children in
that exposure to Mn has cytotoxic effects, including a the same area and repeated 1 year later, it was shown
depressive effect on the number and phagocytic capac- that during the study period of 6 months the recorded
ity of alveolar macrophages (Škreb et al., 1980; Fisher incidence of acute respiratory diseases, as well as the
and Škreb, 1980). Thus, toxicological findings in exper- values of FEVs, correlated in both studies with the lev-
imental animals and in vitro studies have supported els of air-Mn measured at the locations of the schools
human studies and corroborated the hypothesis about attended by the studied children (two schools were
the mechanism of Mn action on the lungs. However, it in zones with higher air-Mn levels, and one in a zone
has to be taken into consideration that an inflamma- with levels of air-Mn about 10 times lower). Acute
tory response is not unique to Mn-containing particles bronchitis was significantly more frequent and FEVs
but is characteristic of nearly all inhalable PM. were lower in children from the schools with higher
air-Mn levels (Hrustić and Šarić 1980).
Bergström (1977) summarized data from epidemio-
7.4.2  Human Studies of Lung Impairment
logical and experimental studies concerning the effect
An increased morbidity and mortality rate from of Mn on the lung. He supported the concept that a pri-
pneumonia had been found among workers exposed mary inflammatory reaction can occur in the lung after
to Mn dusts (Wassermann and Mihail, 1961). Clinically, exposure to MnO2 without the presence of pathogenic
Mn pneumonia has been characterized as an acute bacteria. Such a reaction pattern also explains the inef-
alveolar inflammation with marked dyspnea. A typical fectiveness of the antibiotics commonly used for the
finding was that antibiotics were often without effect. treatment of pneumonia caused by Mn. On the other
996 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

hand, experimental studies suggest that exposure to luteinizing hormone (LH), and prolactin (PRL) (Roels
MnO2 causes a decrease in resistance toward respira- et al., 1992). In a study carried out in three groups of
tory infections. Consequently, some of the observed workers occupationally exposed to Mn (63 miners or
cases of Mn-induced pneumonia might have had a ore processing workers, 38 electric welders in mechani-
bacterial genesis (Šarić and Piasek, 2000). cal fields, and 110 electric welders in shipbuilding), Wu
It has been suggested (Šarić and Lučić-Palaić, 1977) et al. (1996) found increased semen liquefaction time
that long-term exposure to Mn combined with a smok- and decreased sperm count and viability. Although
ing habit may contribute to the development of chronic this study indicates that Mn exposure can cause sperm
nonspecific lung disease. toxicity, the presence of other metals in the welders
In a longitudinal follow-up study on pulmonary (i.e. copper, nickel, chromium, and iron), which were
function and respiratory symptoms in 145 miners also elevated in semen, also had to be taken in con-
exposed to Mn and 65 matched controls (Mashhadi sideration. In animal experiments on rats and rabbits
Abar Boojar and Goodarzi, 2002), the results indicated (Chandra et al., 1973), it was shown that excessive Mn
a synergistic effect of Mn and smoking on the devel- exposure might produce marked degenerative changes
opment of chronic respiratory impairment. Pulmonary in the seminiferous tubules. This effect did not occur
function was also impaired among welders exposed to immediately but developed slowly over the course of
manganese (Bowler et al., 2007). In a study by Halatek 4-8 months following exposure. Direct damage to the
et al. (2005) on neurological and respiratory symptoms testes has not been reported in humans occupation-
in shipyard welders exposed to Mn, it was pointed ally exposed for longer periods. Large amounts of Mn
out that inhibition of Clara cell protein secretion in affect fertility in mammals and are toxic to the embryo
younger welders, i.e. not adapted to the Mn environ- and fetus (Colomina et al., 1996).
ment, suppressed the anti-inflammatory effect in the Reported data reveal conflicting evidence for
respiratory tract and probably enhanced Mn absorp- whether occupational exposure to Mn causes adverse
tion and thus the incidence of subclinical neurotoxic reproductive effects. Noted effects may occur as a sec-
symptoms related to airborne and B-Mn levels. ondary result of neurotoxicity to Mn (ATSDR, 2012). In
a study on ambient Mn exposure and sperm mobility
performed in 200 clients of an infertility clinic, Wirth
7.5  Effects on Other Organs and Systems et al. (2007) showed that a high Mn level was associ-
ated with an increased risk of low sperm mobility and
7.5.1  Reproductive Effects
low sperm concentration. There was a U-shaped dose-
Impotence and loss of libido are common com- response pattern between quartiles of Mn exposure
plaints in workers occupationally exposed to Mn who and all three sperm parameters. In a cross-sectional
have been afflicted with the clinical signs of mangan- study, Ellingsen et al. (2007) compared the serum con-
ism (Emara et al., 1971). Impaired fertility (measured centrations of inhibin B and PRL in 96 male current
as a decrease in the number of children per married welders and 23 patients who were all former weld-
couple) has been observed in male workers exposed ers diagnosed as having welding-related manganism,
for 1-19 years at air-Mn levels that did not produce with 96 age-matched referents. The current welders’
frank signs of manganism (Lauwerys et al., 1985). On GM airborne exposure to Mn was 121 μg/m3 (range
the other hand, in a study performed by Jiang (1996a), 7-2320). PRL level adjusted for age and smoking habits
on 314 men in a Mn plant exposed for up to 35 years to (GM 193 mIU/L vs. 166 mIU/L; P = 0.047) and inhibin
Mn to levels of 0.145 mg/m3 as MnO2 (GM) no signifi- B adjusted for alcohol consumption (arithmetic mean
cant differences were found in reproductive outcomes 151 ng/L vs. 123 ng/L; P = 0.001) were higher in the
between exposed and control workers. However, welders than in referents. The whole-blood Mn con-
impotence and lack of sexual desire were higher in Mn- centration was associated with the serum PRL concen-
exposed workers. In contrast, a study into the reproduc- tration. Tobacco smoking resulted in lower serum PRL
tive effects in 70 male workers of a dry alkaline battery concentrations. The GM of serum PRL concentrations
plant exposed to Mn dioxide at a median concentration of the patients did not significantly differ from that of
of 0.71 mg/m3 in total dust for an average of 6.2 years referents, but the arithmetic mean of serum inhibin B
did not register any difference compared with controls concentrations was significantly lower. These results
(Gennart et al., 1992). Moreover, MnO2-exposed work- suggest an effect of Mn on the pituitary that is revers-
ers in this plant did not show changes in the activity of ible upon cessation of exposure. Lower inhibin B con-
the hypothalamo-pituitary-testicular axis compared to centrations in the patients could point to a functional
controls, as revealed by the measurement of the serum impairment of the testicular Sertoli cells, which may be
concentrations of follicle-stimulating hormone (FSH), caused by a welding fume component.
45 Manganese 997

Since 2000, there have been several studies deal- in Mn milling, smelting, and sintering. The GM of Mn
ing with Mn exposure in pregnancy, the fetus, and the in air was 0.13 mg/m3. The length of exposure varied
newborn. The study of Dorman et al. (2005) deals with from 0 to 35 years. There was no increase in abnor-
the maternal-fetus distribution of Mn in rats following mal electrocardiograms between Mn workers and
inhalation exposure to Mn sulfate. The results of this their matched controls. Arterial blood pressure values
study showed that the placenta partially sequesters showed a greater frequency of low diastolic pressure,
inhaled Mn, thereby limiting exposure to the fetus. In a but this effect was highest in young workers with the
study on B-Mn concentrations and intrauterine growth lowest tenure in the plant. Lee and Kim (2012) recently
(Vigeh et al., 2008), Mn levels in umbilical cord blood showed that B-Mn was associated with an increased
and maternal whole blood were measured in appar- risk of hypertension in data representative of the
ently healthy mothers and their newborns. The study Korean adult population. Different results with respect
suggested that Mn in maternal and umbilical cord to the relationship between Mn and blood pressure
blood might induce different effects on birth weight. may be due to small samples that do not represent a
Another study on maternal B-Mn levels and infant general population or due to different Mn exposure
birth weight (Zota et al., 2009) showed that maternal status and a lack of control for various confounders
B-Mn levels during pregnancy are associated in a non- affecting blood pressure. Therefore, prospective stud-
linear pattern with birth weight in full-term infants. ies are needed to obtain a better understanding of the
Claus Henn et al. (2010) observed an inverted U-shaped role of Mn in hypertension, which may have important
association between 12-month blood manganese and implications for health policy and disease prevention.
concurrent mental development scores. These results Animal studies into the relationship between blood
suggest that an inverted U-shape association between pressure and Mn also showed contradictory results.
B-Mn at 12 months of age and concurrent Mental Kimura et al. (1978) reported that a high dietary expo-
Development Index Score (MDIS), suggesting that both sure to Mn (564 mg/kg) in rats produced a significant
low and high B-Mn levels may have adverse effects on increase in serotonin concentrations and a decrease in
mental development (highest MDIS at a B-Mn level of blood pressure. The authors attributed the final marked
around 25 μg/L, which is a rather high value). The data decrease in blood pressure to the elevated blood sero-
are consistent with manganese being both an essential tonin concentrations, most probably released from dif-
nutrient and a toxicant. Because there is no association ferent tissues.
with B-Mn at 24 months of age, it is possible that 12 Following previous studies suggesting that Mn
months is a sensitive time point for Mn effects on cogni- exposure can be toxic to the heart, Miller et al. (2006)
tion. The same group observed evidence of synergism performed a study on rats. The results showed that
between lead and manganese on neurodevelopmental high dietary Mn reduced mitochondrial O2 consump-
testing in 455 children tested every 6 months between tion. However, dietary Mn, even at high concentra-
12 and 36 months of age, whereby lead toxicity was tions, is unlikely to pose a serious threat to health
increased among children with high manganese coex- when ingested along with adequate dietary Mg.
posure (Claus Henn et al., 2012). In an experimental study (Yang et al., 2007), Mn
cytotoxicity was examined in neonatal rat ventricu-
lar myocytes in vitro by 3-(4,5-dimethythiazol-2-yl)-
7.5.2  Cardiovascular Effects
2,5-diphenyl tetrazolium bromide (commonly known
In an epidemiological study performed in ferroal- as MTT) cell proliferation assays: after incubation with
loy workers, a decrease in systolic blood pressure was different concentrations of Mn for 24 h, apparent cyto-
found (Šarić and Hrustić 1975). Arterial blood pressure toxicity was observed.
was measured and compared in three groups of male
workers aged 20-59 years at different levels of expo-
7.5.3  Hematological Effects
sure to airborne Mn. The lowest mean values of systolic
blood pressure were found in workers with the highest In subjects chronically exposed to high levels of
occupational exposure (0.39-20.44 mg/m3), although Mn in the workplace, no significant hematological
this group was comparatively the oldest. The lowest effects have been observed. From studies into chronic
mean diastolic pressure values were found in control Mn poisoning, it was presumed that large amounts
workers. Confounders such as age, body mass index, of Mn caused depression of both erythropoiesis and
and smoking habits, which might have an impact on granulocyte formation (WHO, 1981). Yiin et al. (1996)
the results obtained, were accounted for. Jiang (1996b) observed increased erythrocyte SOD and plasma malon­
studied the potential cardiotoxicity of MnO2 exposure dialdehyde in males who worked in Mn smelters. In
in 656 workers (547 males and 109 females) engaged the dry alkaline battery study by Roels et al. (1992),
998 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

erythropoietic parameters (red blood cell counts, occupational asthma due to Mn exposure in a 42-year-
hemoglobin, mean corpuscular volume, and mean cor- old nonsmoking welder with work-related dyspnea
puscular hemoglobin concentration) and serum iron (Wittczak et al., 2008) is the first well-documented case
concentration in Mn-exposed workers showed a con- of Mn-induced occupational asthma. The argument for
sistent and statistically significant trend toward lower recognizing the condition as occupational asthma was
values, although the distributions of the values were a positive clinical response to the specific challenge
well within the normal ranges. test, as well as morphological changes found in the
induced sputum.
7.5.4  Endocrine Effects
In a study by Alessio et al. (1989), performed in 7.5.6  Genotoxic and Carcinogenic Effects
foundry workers exposed to Mn levels of 0.04-1.1 mg/m3
Relatively high doses of Mn affect DNA replication
(PM) and 0.5-0.9 mg/m3 as fumes for approximately 10
and repair in bacteria, and cause mutations in both
years, elevated PRL and cortisol levels were reported.
microorganism and mammalian cells, although the
No changes in the levels of FSH and LH were noted.
Ames test does not appear to be particularly respon-
Except for serum PRL, a similar observation was
sive to Mn. In mammalian cells, Mn causes DNA dam-
made by Roels et al. (1992) for serum FSH and LH in
age and chromosome aberrations (Gerber et al., 2002).
dry alkaline battery workers exposed to MnO2 air-
In a more recent study performed by Lima et al. (2008)
borne dust. Smargiassi and Mutti (1999) reported also
with the aim of evaluating the mutagenic potential of
an increase of serum PRL levels in a group of work-
Mn chloride, the comet assay and chromosome aber-
ers from a ferroalloy plant. Measurements were made
ration analysis were applied to determine the DNA-
twice in the same workers (in 1992 and 1997). Serum
damaging and clastogenic effects of MnCl2.4H2O.
PRL levels that were significantly elevated in the ear-
Cultured human lymphocytes were treated with
lier analysis (i.e. 1992) had increased significantly
15, 20, and 25 μM Mn chloride. MnCl2.4H2O showed
during the intervening period in 1997. During the
strong cytotoxicity at all phases of the cell cycle. Both
5-year period between studies, exposure levels were
genotoxicity observed at the G2 phase and the comet
consistent.
assay may reflect the lack of time for cellular repair
Pine et al. (2005) performed an experimental study
system to act. The absence of chromosome aberrations
on female rats to determine the effects of Mn exposure
at all other phases of the cell cycle suggests that Mn-
on puberty-related hormones and the onset of female
mediated damage may be repaired in vitro.
puberty. MnCl2 administered acutely into the third
In another study published in the same year (Jiao
ventricle of the brain acted in a dose-dependent man-
et al., 2008), the authors examined single-strand breaks
ner to stimulate LH release in prepubertal female rats.
(SSBs) in mitochondrial DNA (mtDNA) induced by
These results suggest that Mn may contribute to preco-
MnCl2 in vitro and in vivo and discuss the possible
cious puberty if an individual is exposed to elevated
underlying mechanism. In vitro results indicated that
levels of Mn at an early stage in their development.
MnCl2 increased the formation of mtDNA SSBs and ROS
in hepatic mitochondria in a dose-dependent manner.
7.5.5  Immunological Effects
MnCl2 exposure in vivo increased mtDNA SSBs in rat
In a study performed in male welders exposed to brain and liver, and decreased the level of glutathione
Mn (0.29-0.64 mg/m3) for an unspecified duration, in rat hepatic mitochondria and brain homogenates in
suppression of T and B lymphocytes, characterized a dose-dependent manner. The level of malondialde-
by reductions in serum immunoglobulin G and total hyde and the activities of SOD and glutathione per-
E rosette-forming cells were observed (Boshnakova oxidase (GPx) were not significantly changed in both
et al., 1989). However, the welders in this study were hepatic mitochondria and brain homogenates in rats.
also exposed to other compounds, including cobalt, These results indicate that Mn treatment increases the
carbon dioxide, and nitric acid, as well as to noise and amount of mtDNA SSBs in vitro and in vivo, probably
vibration. In addition, it is not known whether any of mediated by Mn-induced oxidative stress.
the registered changes are associated with significant The fungicide maneb and the contrasting agent
impairment of immunological system function. MnDPDP can be embryotoxic, but for MnDPDP the
Manganese belongs to a group of agents called effect only occurs at doses much higher than those
transition metals, together with chromium, cobalt, employed clinically (Maci and Arias, 1987).
and nickel, which are known to induce occupational There is only one human study into the genotoxic
asthma. The similar chemical structure of Mn may thus effects of exposure to Mn. In this study, an increased
determine its asthmogenic potential. A case report on incidence of chromosomal aberrations was registered
45 Manganese 999

in three groups of welders with occupational expo- The RfC is intended to be the air manganese concentra-
sures lasting 10-26 years (Elias et al., 1989). In addition tion to which the population can be exposed for a life-
to Mn exposure (median concentrations of 0.18 mg/m3 time without appreciable risk of adverse health effects.
for respirable dust and 0.71 mg/m3 for total dust), the The United States Agency for Toxic Substances and
welders were also exposed to nickel and chromium. Disease Registry recently published a new “minimal
Therefore, the observed increase in chromosomal risk level” for respirable Mn of 0.3 μg/m3 for chronic
aberrations could not be attributed to exposure to any exposure inhalation for 365 days (ATSDR, 2012).
single metal (nickel is known to cause chromosomal For occupational exposure, the ACGIH currently
aberrations by the inhalation route). adopts for inhalable Mn particles a TLV-TWA of 0.2 mg
In a study into the genotoxic effects of occupational Mn/m3 (listed as elemental Mn and inorganic com-
exposure to mancozeb (in 14 women and 30 men, all pounds, as Mn) and a 0.02 mg Mn/m3 TLV-TWA for
of reproductive age), Jablonickà et al. (1989) showed a respirable particles with a notation that the TLV is
significantly increased incidence of chromatid breaks based on “CNS func” (i.e. neurobehavioral and neuro-
in exposed women compared to controls, and in the psychological changes) (ACGIH, 2013). The U.S. Occu-
combined exposed cohort compared to the combined pational Safety and Health Administration (OSHA,
control group. Isochromatid breaks were significantly 1998) adopted in 1998 0.2 mg/m3 as the permissible
increased in exposed men and in the combined group, exposure limit (PEL)-TWA for Mn, both elemental and
compared to their respective controls. Gaps were inorganic compounds. The earlier OSHA PEL value of
significantly increased in both exposed groups. The 5 mg Mn/m3 is still valid as the ceiling value.
number of sister chromatid exchanges (SCEs) was The upper range of the ESADDI (i.e. 5 mg/day)
not significantly increased in any group. Mancozeb- (NAS/NRC, 1989) was indicated as a provisional
exposed smokers had a significantly increased inci- guidance value (0.07 mg/kg/day) for oral exposure
dence of SCEs compared to exposed nonsmokers. to Mn. The EPA has derived a chronic oral reference
Information on cancer due to Mn is scanty, but the dose (RfD) of 0.14 mg/kg/day for Mn. This value cor-
available data do not indicate that inorganic Mn is car- responds to the average daily intake of Mn in the diet
cinogenic. The EPA (1996) has concluded that Mn is not considered to be adequate and safe (10 mg/day). The
classifiable as a human carcinogen. The International RfD was derived assuming an average body weight of
Agency for Research on Cancer has not evaluated Mn 70 kg and was based on a composite of data provided
(Boffetta, 1993). So far, there is insufficient evidence to by the WHO and the NRC. An uncertainty factor was
indicate that inorganic Mn exposure produces cancer not employed for the following reasons: the informa-
in animals or humans. tion used to determine the RfD was taken from many
large populations; humans exert efficient homeostatic
control over Mn such that body burdens are kept con-
8  GUIDELINES AND REGULATIONS stant with variations in diet; there are not subpopula-
tions that are believed to be more sensitive to Mn at this
The WHO Regional Office for Europe indicated a level; Mn is an essential element that is required for
guideline value for environmental air-Mn of 0.15 μg/m3 normal human growth and the maintenance of health
(WHO, 1994). Based on early neurotoxic effects (ATSDR, 2012). However the EPA has recommended
observed in occupationally exposed workers and that a “modifying factor” of three should be applied
using the benchmark approach, an estimated no when assessing risk from Mn in drinking water or soil
observed adverse effect level [or NOAEL; the lower because the study by Kondakis et al. (1989) raises sig-
95% confidence limit of the benchmark dose lower nificant concerns about possible adverse neurological
limit (BMDL5) for a 5% excess in the background prev- effects at doses not far from the range of essentiality
alence] of 30 μg/m3 was obtained. The guideline value (ATSDR, 2012).
should be applied as an annual average. The EPA pub- For drinking water, the WHO issued a guideline
lished a reference concentration (RfC) of 0.05 μg Mn/ of 400 μg/L for Mn in the third edition of “Guidelines
m3 (EPA, 2012a), based on the lowest observed adverse for Drinking-Water Quality” (WHO, 2004a). However,
effect level of 0.15  mg/m3 from the occupational the 400-μg/L guideline for Mn was discontinued in
study by Roels et al. (1992) and conducted a reevalu- the fourth edition because “this health-based value
ation that recommended a range of 0.09-0.20 μg Mn/ [400 μg/L] is well above concentrations of Mn nor-
m3, although the RfC was not formally changed (EPA, mally found in drinking-water, [so] it is not considered
1994). Health Canada (2010) has reviewed the health necessary to derive a formal guideline value” (WHO,
effects of manganese and derived a RfC of 0.05 μg/m3 2011). A review of WHO publications, peer-reviewed
from the occupational study by Lucchini et al. (1999). journal articles, government reports, and published
1000 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

conference proceedings strongly suggests that Mn A recent study by Kim and Lee (2011) showed that
levels > 400 μg/L in drinking water or in potential women had a higher GM of B-Mn concentrations
drinking-water supplies are found in a substantial than men in a representative Korean general adult
number of countries (Frisbie et al., 2012). In addition, population (14.03 μg/L vs. 11.92 μg/L). This difference
drinking mineral water regularly can add significantly is mainly due to differences in iron status between
to Mn intake (Dieter et al., 1992). Recent research into men and women (Baldwin et al., 1999; Aschner and
the health effects of Mn on children suggests that the Aschner, 2005; Meltzer et al., 2010) because iron defi-
earlier WHO guideline of 400 μg/L was too high to ciency increases B-Mn level and the prevalence of iron
adequately protect public health. deficiency in women is higher than in men (Davis,
C.D., et al., 1992; Erikson et al., 2002a, Kim and Lee,
2011). However, the study by Kim and Lee (2011) also
9  BIOMARKERS OF EXPOSURE AND showed that women with normal hemoglobin or fer-
EFFECTS ritin levels nevertheless had significantly higher B-Mn
concentrations than men. The difference in B-Mn even
9.1  Mn Concentration in Biological Media after controlling for iron deficiency status may be due
to gender differences instead of differences in ferritin
as a Biomarker of Exposure
level (Kim and Lee, 2011). Norwegian women with
Being an essential element, Mn is actively controlled serum ferritin concentrations ≥ 12 μg/L and hemo-
by homeostatic mechanisms regulating its absorption, globin ≥ 12.0 g/dL (120g/L) had average B-Mn con-
distribution, and excretion. These processes also play centrations of 9.7 μg/L (Meltzer et al., 2010), whereas
an important role in Mn toxicokinetics, in contrast to Norwegian men on average had blood concentrations
many nonessential toxic metals such as mercury and of 8.8 μg Mn/L at a normal mean serum ferritin level.
lead. As a result of this highly efficient control, the Ellingsen et al. (2003b) observed a lower mean concen-
association between external and internal exposure tration of soluble TfR in Mn-exposed workers than in
indicators is weak at the individual level. Several bio- controls (2.2 vs. 2.6 mg/L; P < 0.001), and the concen-
markers are used and others are being investigated tration was negatively associated with current expo-
in an attempt to develop accurate indicators to reflect sure to “soluble” Mn in the inhalable aerosol fraction
the body burden or current exposure. Monitoring con- and with current smoking habits. They also observed
centrations in body fluids can identify individuals an association between the concentration of serum-sol-
with excessive exposure and those at risk of develop- uble TfR and the concentration of Mn in whole blood
ing clinical abnormalities (Lucchini and Kim, 2009). (Pearson’s r = 0.48; P < 0.001) in controls. According to
Manganese can be measured with good sensitivity the authors, these results suggest that Mn-exposed
in biological fluids and tissues, and levels in blood, workers have higher intracellular iron concentration in
urine, feces, and hair have been investigated as pos- erythrocyte precursors compared to controls, resulting
sible biomarkers of exposure. The reported normal in downregulation of TfRs on the surface of these cells.
concentrations of Mn in blood, serum, and urine show A recent study showed that premenopausal women
a wide range. Data by Buchet et al. (1976), Zielhuis had on average 1.5 μg/L higher B-Mn concentrations
et al. (1978), Baldwin et al. (1999), and Kim and Lee than did postmenopausal women, mainly due to dif-
(2011) using electrothermal atomic absorption indi- ferences in iron status between premenopausal and
cate that the average normal level in whole blood is postmenopausal women (Lee and Kim, 2012).
7-13 μg/L. Approximately 85% of B-Mn is in erythro- Baldwin et al. (1999) also reported that GMs for Mn
cytes; for serum or plasma, the average Mn concentra- levels were higher in women than in men (7.50 μg/L
tions are reported to be 0.6-1.0 μg/L, as determined versus 6.75 μg/L). In addition, for women, Mn blood
by electrothermal atomic absorption (Halls and Fell, levels correlated with serum iron and decreased as age
1981). Milne et al. (1990) reported the mean Mn contri- increased.
bution of blood components to whole blood (and per- Manganese levels in blood increase in pregnant
centage of total Mn) for healthy adult Americans (24 women, with levels at full term measured to be at least
men, 24 women; 25-49 years of age): for whole blood, twice as high as in nonpregnant women, while cord
10.9 μg/L (100%); for red blood cells, 7.39 μg/L (65.8%); B-Mn levels were approximately twice as high as mater-
for plasma, 0.49 μg/L (4.4%); for white blood cells, nal levels (Smargiassi et al., 2002; Takser et al., 2004; Zota
2.60 μg/L (23.2%); and for platelets, 0.74 μg/L (6.6%). et al., 2009). Infants have higher levels of B-Mn compared
In healthy, unexposed people living in the Lombardy to children or adults (Rukgauer et al., 1997; Rice et al.,
region in northern Italy, B-Mn was 8.8 ± 0.2 μg/L and 2010). The increase in Mn is mainly due to increased
serum Mn was 0.6 ± 0.014 μg/L (Minoia et al., 1990). needs for iron in pregnancy, the fetus, and infants.
45 Manganese 1001

Although urinary excretion of Mn is low (less than of measuring Mn in blood and urine as a measure of
1% of the absorbed dose), measurements of urine con- excess Mn exposure is the relatively rapid rate of Mn
centrations have been made. The reported normal con- clearance from the blood circulation and the very low
centrations of Mn in urine vary to a large extent. Hoet excretion rate in urine. Manganese excreted in urine
et al. (2013) recently performed a nationwide survey represents only a small fraction of eliminated Mn, and
to determine (using ICP-MS) the reference urinary Mn the variability of Mn in urine is high among exposed
concentration in the general Belgian population (460 and even among control subjects; therefore, Mn in
men, 514 women; age 18-80 years). The P5-P95 urinary urine is less useful than Mn in blood as an exposure
Mn concentration interval in men and women (n = 974) biomarker at the group level (Lucchini and Kim, 2009;
combined amounted from < 0.043 to 0.36 μg/L; the Zheng et al., 2011).
arithmetic mean was < 0.043 μg/L (range < 0.043 to In a Canadian study by Baldwin et al. (1999) into the
1.72 μg/L). Earlier data by Buchet et al. (1976), Halls relationship between levels of Mn in the air and in blood,
and Fell (1981), Oberdorster and Cherian (1988), and significantly higher levels of B-Mn correlated with high
Minoia et al. (1990) corroborate the Hoet et al. (2013) levels of airborne Mn. Data obtained in this study com-
findings in that they indicate that less than 1 μg Mn/L bined with the occupational studies indicate that there
is normally found in the urine of humans never occu- may be a plateau level related to homeostatic control of
pationally exposed to Mn. Mn on an individual basis. At low levels, B-Mn concen-
On a group basis, workers exposed to a mean con- trations would be related to food, water, and air levels
centration of 1 mg Mn/m3 had higher levels of Mn in of Mn, and large differences in individual blood levels
whole blood and urine compared to unexposed con- would be observed. At high exposure levels, such as in
trols (Roels et al., 1987b); the group average levels of occupational environments, this plateau may be reached
whole B-Mn appear to be related to Mn body burden or exceeded. A study by Hoet et al. (2013) in 27 Mn-
and the urine Mn levels to current exposure. However, exposed welders (air-Mn 1.3 to 729 μg/m3; GM 28 μg/m3)
the individual measurements of blood or urine Mn do showed after the first shift of the working week (thus,
not correspond to individual external exposure levels after 2 days of nonexposure), values of Mn in plasma
(Roels et al., 1987a,b, 1992; Zheng et al., 2011). (P-Mn) that correlated well on an individual basis with
The study by Lucchini et al. (1995) suggests that air-Mn levels above 10 μg/m3 (n = 20; r = 0.69; P < 0.001).
blood and urine levels are correlated with exposure At the end of the second working day, the slope of the
levels on individual basis. In this study, a correlation P-Mn values vs. air-Mn dropped 2.3 times because of the
was observed between a cumulative exposure index increased response of the homeostatic system to main-
(CEI) and B-Mn, and both B-Mn and CEI were asso- tain “normal” systemic Mn levels. A P-Mn cutoff value of
ciated with neurobehavioral outcomes. Workers were 2 μg/L can distinguish air-Mn above or below 20 μg/m3
tested during a month of being laid off, and all cor- with a sensitivity of 69% and a specificity of 82%.
relations increased proportionally to the period from The use of hair as a possible biomarker of Mn expo-
the exposure cessation. Mergler and Baldwin (1997) sure is problematic. In hair, Mn concentrations are
suggested that in this study the correlation between reported to be normally below 4 mg/kg (Oberdorster
CEI and B-Mn was evident because B-Mn levels were and Cherian, 1988). While some studies have found a
not influenced by current exposure and were there- correlation between exposure levels and Mn concen-
fore better related to the Mn levels accumulated in the tration in the hair (Collipp et al., 1983; Huang and Cao,
body storage deposits. This situation has not yet been 2003; Lin, 2002; Wang and Wu, 2000), the majority of
reproduced in the literature. In a follow-up study of studies reported different limits concerning the valid-
currently exposed workers, Lucchini et al. (1999) did ity of scalp hair analysis as a biomarker of exposure
not observe any correlation between CEI and B-Mn, (Lyden et al., 1984; Stauber et al., 1987; Sturaro et al.,
although the CEI was still associated with the impair- 1994). A more rigorous hair cleaning methodology was
ment of neurobehavioral functions. developed by Eastman et al. (2013) to establish the
Other studies on currently exposed workers by Jär- utility of hair as an exposure biomarker for Mn and
visalo et al. (1992) and Roels et al. (1987a,b, 1992) indi- other metals (Pb, Cr, Cu), using ICP-MS, scanning elec-
cate that on an individual basis, correlations between tron microscopy, and laser-ablation ICP-MS to evalu-
the level of workplace air-Mn exposure and the lev- ate cleaning efficacy. The cleaning method included
els of Mn in blood and urine are not reliable predic- 0.5%Triton X-100 sonication plus 1 N nitric acid soni-
tors of external Mn exposure. Järvisalo et al. (1992), cation and was highly effective in removing dust
as well as Roels et al. (1987a,b, 1992), suggested that contamination from hair samples from adolescents.
blood and urinary Mn levels may be useful to moni- Manganese hair concentration was ∼4-70-fold lower
tor group exposure. A factor that limits the usefulness than levels reported in other pediatric Mn studies and
1002 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

were significantly higher in the proximity of ferroalloy branch from the bronchioles. CC16 damage causes a
emissions. significant change in the levels of this protein. The pro-
Given the complex and limited relationship between tein can be quantified in serum and urine. Until now,
exposure and B-Mn levels may depend upon exposure the protein has only been studied experimentally fol-
attributes and the latency of blood sampling relative to lowing i.p. administration of MMT.
exposure and the scarce utility of plasma and urine as Manganese exposure is known to alter iron (Fe)
exposure biomarkers (Smith et al., 2007), other candi- homeostasis. Zheng et al. (2011) showed that the
date biomarkers are being measured in different occu- plasma or erythrocyte Mn:Fe ratio reflects not only
pational and environmental settings: tooth enamels steady-state concentrations of Mn or Fe in tested indi-
(Ericson et al., 2007), nails (Wongwit et al., 2004; Gil viduals but also a biological response (i.e. altered Fe
et al., 2011; Laohaudomchok et al., 2011; Sriram et al., homeostasis) to Mn exposure; the ratio may be a sen-
2012), and saliva (Gil et al., 2011; Wang et al., 2008). sitive measure of Mn exposure. However, the cutoff
Particularly relevant is the possibility of measuring values for plasma or erythrocyte Mn:Fe ratio among
Mn in teeth using laser-ablation ICP-MS. This new the general population needs to be further explored,
technique allows the determination of pre- and post- verified, and established.
natal exposure, as well as a cumulative early life expo- Serum PRL has been identified as a possible bio-
sure. Higher Mn levels in prenatal dentin provided marker of Mn effects on dopamine neurotransmis-
retrospective and time-specific levels of fetal expo- sion (Montes et al., 2011; Marreilha Dos Santos et al.,
sure resulting from environmental and occupational 2011; Aschner, 2006b; Takser et al., 2004); namely, the
sources in the community of Salinas, California, one of tuberoinfundibular dopaminergic system, which
the areas with the highest use of Mn-containing fungi- exerts tonic inhibition of PRL secretion, is affected by
cides (Gunier et al., 2013). Mn dust loading (μg/m2 of Mn action (Smargiassi and Mutti, 1999). Still, it is not
floor area) during pregnancy were associated with the clear whether PRL levels indicate recent or cumulative
possibility of determining fetal levels from the mantle exposure. It should also be mentioned that in studies
dentine in deciduous teeth (Arora et al., 2012). In many by Roels et al. (1992) and Myers et al. (2003b) serum
ways, animal studies have been highly productive for PRL levels were not increased in workers chronically
understanding the mechanism of toxicity and for dif- exposed to airborne Mn.
ferentiating the impact of different exposure windows
in relation to cumulative lifelong exposure. Prewean-
ing Mn exposure caused hyperactivity, disinhibition, 9.3 Neuroimaging
and spatial learning and memory deficits associated Tremendous progress has been made in medical
with altered D1 and D2 DARs and DAT levels (Kern imaging of the human body since the invention of com-
et al., 2010). The results of different tests for motor puted tomography (CT) and MRI. Neuroimaging of
skills were differentially impacted by early life and/or patients with metal neurotoxicity can be divided into
lifelong exposure through oral administration of Mn two types: morphological neuroimaging (anatomy-
(Beaudin et al., 2013). based imaging), including CT and MRI; and functional
neuroimaging (physiology-based imaging), such as
magnetic resonance spectroscopy (MRS), single-pho-
9.2  Other Biomarkers
ton emission computed tomography (SPECT), PET,
Lymphocyte Mn-dependent SOD activity increases diffusion tensor imaging (DTI), and functional MRI
with increased Mn uptake (Yiin et al., 1996). As sug- (fMRI). Neuroimaging is undergoing a shift from mor-
gested by the results of some studies (Davis and Gre- phological to functional imaging as new technologies
ger, 1992; Greger, 1999), the analysis of this enzyme, are introduced (Lang, 2000; Walker et al., 2004). MRI,
in conjunction with serum Mn levels, may be used in PET, and SPECT have been used for 10 years or more
assessing low and moderate levels of exposure to Mn. to evaluate workers exposed to Mn, and to examine the
Ethylene thiourea is readily detected in the urine neurological consequences of such exposure. Recently,
of humans (and animals) exposed to maneb and man- functional neuroimaging modalities such as fMRI,
cozeb, but there is no clear relationship between dose MRS, and DTI have also been applied (Kim, 2011).
and urinary levels (Kurttio et al., 1990; Kurttio and
Savolainen, 1990).
9.3.1  Pallidal Signal Intensity on T1-Weighted
Clara cell secretory protein CC16 is a potential
Magnetic Resonance Imaging
biomarker for the pulmonary effects of exposure to
MMT (Halatek et al., 1998). Clara cells are unciliated The paramagnetic properties of Mn cause shortening
cells that occur at the boundary where alveolar ducts of the nuclear magnetic resonance proton T1 relaxation
45 Manganese 1003

times and signal hyperintensity on T1-weighted brain the Mn source has been shown after cessation of occu-
MRI. A Mn-induced bilateral symmetrical increase pational exposure (Nelson et al., 1993), discontinuation
in signal intensities, confined to the globus pallidus of TPN (Mirowitz et al., 1991), and liver transplanta-
and midbrain, can be observed on T1-weighted MRI, tion in patients with hepatic failure (Choi et al., 2005;
but with no alteration to the T2-weighted images Pujol et al., 1993). These findings suggest that increased
(Kim, 2004). However, Mn-induced high signals on signal intensities on a T1-weighted image reflect expo-
T1-weighted MRI do not correspond to any abnormal sure to Mn, but do not necessarily indicate the presence
findings on brain CT (Park et al., 2003). The characteris- of manganism. Many reports have shown that B-Mn
tic high signal caused by Mn can be differentiated from is highly correlated with PI in liver cirrhotics (Hauser
signals that increase in intensity for other reasons. Thus, et al., 1996; Krieger et al., 1995; Spahr et al., 1996). In
high signals from fat, hemoglobin breakdown products, Mn-exposed workers, B-Mn was also found to correlate
melanomas, neurofibromatosis, and calcification can be with PI (Chang et al., 2009a; Jiang et al., 2007; Kim et al.,
seen on T1-weighted images. High signals from hemo- 1999a).
globin breakdown products, melanomas, and neurofi- A recent study showed that PI was significantly
bromatosis can be differentiated from Mn-induced high associated with digit symbol test results, digit span
signals on the basis of signal site and symmetry. Iron backward ratings, scores on the Stroop Word and
deposits cause a greater shortening of the T2-relaxation Stroop Error indices, and grooved pegboard (domi-
time than the T1-relaxation time, resulting in low signal nant hand) data (Chang et al., 2009a ). This means that
intensity upon T2-weighted imaging, which is distinct PI is a good predictor of neurobehavioral performance
from that of an Mn deposit (Kim, 2004). Calcification in welders without clinical manganism. In particular,
can be easily identified by CT (Ahn et al., 2003). Krieger PI was a better predictor than B-Mn levels of neurobe-
et al. (1995) coined the term pallidal index (PI) to quan- havioral performance in such welders (Chang et al.,
tify Mn accumulation in the globus pallidus, defined as 2009a).
the ratio of the signal intensity in the globus pallidus Taken together, the data suggest that the appear-
to that in the subcortical frontal white matter (WM) in ance of PI on MRI may reflect a target organ dose of
axial T1-weighted MRI planes, multiplied by 100. An occupational Mn exposure (Kim, 2006). In addition,
increase in signal upon T1-weighted imaging has been Mn in the brain has a longer half-life than in the blood
shown in experimental Mn intoxication of rats (Gallez (Lucchini and Kim, 2009). Thus, PI reflects the cumula-
et al., 2001) and nonhuman primates (Erikson et al., tive dose better than the B-Mn level. However, there
1992a; Newland et al., 1989; Dorman et al., 2006), and are some limitations. While the PI is a valid measure
in persons with occupational Mn intoxication (Nelson for comparing MRI images of subjects within a single
et al., 1993; Kim, 2004: Chang et al., 2009b). A similar study (using the same scanner, MRI sequence param-
MRI pattern has also been observed in patients receiv- eters, coils, etc.), its numerical value may depend on
ing total parenteral nutrition (TPN) by direct intrave- the exact imaging parameters, as well as on the com-
nous Mn administration (Ejima et al., 1992; Mirowitz parison region of the brain used for the denominator
et al., 1991) and in patients with portal systemic shunts in the PI (Guilarte et al., 2006). Exact numerical values
such as occur in individuals with liver cirrhosis, lead- of PI may therefore vary between studies (Zheng et al.,
ing to an inability to clear Mn via biliary excretion (But- 2011). Further, the level of signal intensity indicating
terworth et al., 1995; Butterworth, 2010; Hauser et al., progression to manganism from Mn exposure remains
1994, 1996; Krieger et al., 1995; Park et al., 2003; Spahr to be determined.
et al., 1996). Manganese-induced high signals can
occasionally be observed in patients with severe iron-
9.3.2  PET/SPECT Indicate the Integrity of the
deficiency anemia (Kim et al., 2005). Kim et al. (1999a)
Dopaminergic Nigrostriatal Pathway
showed for the first time that the characteristic high T1
signals were also frequently observed in asymptomatic In nonhuman primates and humans intoxicated
workers exposed to Mn. The authors found a high prev- with Mn, 18F-dopa PET scans are normal (Erikson
alence (41.6%) of increased MRI signals in Mn-exposed et al., 1992a; Kim et al., 1998; Shinotoh et al., 1995, 1997;
workers. High MRI signals also have been successfully Wolters et al., 1989), whereas in PD, dopamine uptake
used to identify Mn accumulation in children exposed is reduced in the striatum, particularly the posterior
to excess Mn (Devenyi et al., 1994; Fell et al., 1996; Ono putamen, thus resulting in reduced F-dopa uptake in
et al., 1995). The increased signal intensities resolved PET. This supports the view that in Mn intoxication
significantly approximately 1 year after Mn exposure the nigrostriatal pathway is relatively well preserved,
ceased (Kim et al., 1999b). The disappearance of high which is consistent with many pathological obser-
signal abnormalities on MRI following withdrawal of vations showing that Mn-induced damage occurs
1004 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

primarily in pathways postsynaptic to the nigrostria- Using [1H]-MRS, Chang et al. (2009b) showed that the
tal system (Kim, 2006; Guilarte, 2010). DAT imaging ratios of N-acetylaspartate (NAA):total creatine (tCr),
using (1r)-2b-carboxymethoxy-3b-(4-iodophenyl) tro- Glx including both glutamine and glutamate (Glx):tCr,
pane (β-CIT), employed as a SPECT ligand, reveals and total choline (tCho):tCr in both the anterior cin-
the DAT density, and therefore explores the integrity gulate cortex (ACC) and parietal WM did not differ
of the nigrostriatal dopaminergic system. In PD, 123I-β- significantly between welders and controls. However,
CIT SPECT reveals that specific striatal β-CIT uptake myoinositol (mI) levels in the ACC, but not in parietal
is reduced (Seibyl et al., 1995). However, striatal DAT WM, were significantly lower in welders compared to
uptake is nearly normal in patients with Mn-induced controls. Further, in the frontal brain lobe the mI:tCr
parkinsonism (Huang et al., 2003). Various ligands that ratio was significantly positively correlated with ver-
bind to DAT, such as 123I-β-CIT, 123I-fluoropropyl-CIT, bal memory scores and negatively with blood-Mn. The
and 99mTc-TRODAT-1, have been used in SPECT stud- authors suggested that reduction of the mI:tCr in weld-
ies (Huang et al., 2003; Kim et al., 2002). DAT SPECT is ers may reflect a possible glial cell effect rather than
more easily accessible and less expensive than F-dopa a neuronal effect, and may be associated with long-
PET. Racette et al. (2005a) found relatively symmetri- term exposure to Mn. These results agree with those
cal and severely reduced F-dopa uptake on PET in the of a previous study by Kim et al. (2007), who found
posterior putamen of a patient with manganism sec- no significant differences in the levels of brain metabo-
ondary to liver failure, together with T1 hyperintensi- lites (only NAA and Cho were measured) between
ties in the basal ganglia on MRI. Criswell et al. (2012a) welders and controls, although a direct comparison
also reported a reduced striatal F-dopa uptake on PET of mI levels was not possible. However, Guilarte et al.
in a patient with manganism secondary to liver failure, (2006) reported a reduction in the NAA level in the
together with T1 hyperintensities in the basal ganglia parietal cortex and frontal WM in the brains of Mn-
on MRI. However, SPECT data from secondary man- exposed monkeys. Recently, Dydak et al. (2011) used
ganism patients (Kim et al., 2010) revealed two differ- [1H]-MRS with the MEGA-PRESS (i.e. point-resolved
ent patterns of clinical and neuroradiological features. spectroscopic sequence) to determine GABA levels in
Four out of five patients showed atypical parkinson- the thalamus of Mn-exposed smelters. They showed a
ism with normal DAT density, which could be clearly significant decrease in the NAA:Cr ratio in the frontal
differentiated from PD, whereas one patient showed cortex and a significant increase of GABA level in the
l-dopa-responsive parkinsonism with reduced DAT thalamus of exposed subjects. Further studies using
density (classical PD). Further work with PET scan- MRS are needed to identify brain metabolites in Mn-
ning is needed to clarify whether Mn damage can also exposed workers.
extend to the nigrostriatal pathway, as suggested by Chang et al. (2010a) performed the first fMRI experi-
Racette et al. (2005a). ment, using sequential finger tapping, to investigate
Some welders showed clinical features and PET/ the behavioral significance of additionally recruited
SPECT findings typical of PD, with concurrent high brain regions in welders who had experienced chronic
pallidal signals and increased B-Mn (Kim et al., 1999b, Mn exposure. Their findings suggest that when rela-
2002; Racette et al., 2001). Recently, active, asymptom- tively simple tasks are set, motor fMRI may uncover
atic welders with Mn exposure demonstrated reduced evidence of compromised brain function in patients
caudate F-dopa PET, which appeared to be distinct with subclinical manganism. The finding of excessive
from the pattern of dysfunction found in symptomatic recruitment of the cortical motor network in chroni-
PD (Criswell et al., 2011). Kim et al. (1999b) suggested cally Mn-exposed group is in line with the emerging
that Mn might have been a risk factor for the develop- concept of the use of adaptive neural mechanisms to
ment of PD, although they could not exclude the pos- compensate for latent dysfunction in the basal ganglia
sibility that the patient simply suffered from PD, with (Buhmann et al., 2005).
coincident exposure to Mn (Kim et al., 2002). Neuro- Chang et al. (2010b) also performed the first-ever
imaging modalities such as MRI and PET/SPECT may fMRI combined with 2-back memory tests to assess the
therefore be useful for the differential diagnosis of par- neural correlates of Mn-induced memory impairment
kinsonism (Calne et al., 1994; Kim, 2006). in response to subclinical dysfunction in the working
memory networks of welders exposed to Mn chroni-
cally. fMRI with a 2-back memory task indicated that
9.3.3  Functional Neuroimaging
Mn-exposed welders are likely to require more neural
In vivo proton MRS ([1H]-MRS) is an image-guided, resources in working memory networks to compen-
noninvasive method of monitoring neurochemical sate for subtle deficits in working memory and altered
metabolites in the brain (Rosen and Lenkinski, 2007). working memory processes.
45 Manganese 1005

DTI is a unique method used to characterize WM Ballatori, N., Miles, E., Clarkson, T.W., 1987. Am. J. Physiol. 252 (5 Pt 2),
microintegrity, and relies on the principle that water R842–R847.
Baranyi, M., Milusheva, E., Vizi, E.S., et al., 2006. J. Chromatogr. A
diffusion is highly anisotropic in brain WM structures 1120, 13–20.
(Beaulieu, 2002). Direct comparison between 30 welders Barbeau, A., 1984. Neurotoxicology 5, 13–35.
and 19 controls using investigator-independent statistical Beaudin, S.A., Nisam, S., Smith, D.R., 2013. Neurotoxicol. Teratol.
parametric mapping voxel-wise analysis of DTI metrics 38, 36–45.
revealed in Mn-exposed welders a reduction in fractional Beaulieu, C., 2002. NMR Biomed. 15, 435–455.
Becker, J.S., Dietze, H.J., 2000. Fresenius. J. Anal. Chem. 368, 23–30.
anisotropy (FA) in the genu, body, and splenium of the Beklemishev, M.K., Stoyan, T.A., Dolmanova, I.F., 1997. Analyst 122,
corpus callosum (CC), and in the frontal WM (Kim et al., 1161–1165.
2011b). Further, marked increases in radial diffusivity Bergström, R., 1977. Scand. J. Work Environ. Health 3 (Suppl. 1),
(RD), but negligible changes in axial diffusivity, were evi- 1–40.
dent in the genu, body, and splenium of the CC, and in Bernardino, B.E., Young, S.W., Lee, J.K., et al., 1992. Radiology 183,
53–58.
the frontal WM. These findings suggest that microstruc- Blumberg, K., Walsh, M.P., 2004. Status Report Concerning the Use
tural changes such as a reduction of FA with increases in of MMT in Gasoline. http://www.healthandcleanair.org/mmt/
RD in the CC and frontal WM result from a compromised icct.pdf (accessed 27.01.14).
radial directionality of fibers in such areas, primarily Boffetta, P., 1993. Scand. J. Work Environ. Health 19 (Suppl. 1), 67–70.
caused by demyelination. The correlations between FA in Bonilla, E., 1978. J. Neurochem. 31, 551–552.
Bonilla, E., 1984. Exp. Neurol. 84, 696–700.
the frontal WM and the Stroop test scores suggest that Boojar, M.M., Goodarzi, F., 2002. J. Occup. Environ. Med. 44,
poor performance of executive function is closely associ- 282–290.
ated with lower FA values in the frontal WM. Borgese, L., Salmistraro, M., Gianoncelli, A., et al., 2012. Talanta 89,
99–104.
Boshnakova, E., Divanyan, H., Zlatarov, I., et al., 1989. J. Hyg. Epide-
miol. Microbiol. Immunol. 33, 379–382.
References Bouchard, M., Laforest, F., Vandelac, L., et al., 2007. Environ. Health
Perspect. 115 (1), 122–127.
ACGIH (American Conference of Industrial Hygienists), 1995. 1995 Bouchard, M.F., Sauvé, S., Barbeau, B., et al., 2011. Environ. Health
TLVs and BEIs. ACGIH, Cincinnati, OH, USA. Perspect. 119 (1), 138–143.
ACGIH, 1999. Particle size-selective sampling for particulate Bowler, R.M., Roels, H.A., Nakagawa, S., et al., 2007. Occup. Envi-
air contaminants. In: Vincent, J.H. (Ed.), ACGIH, Cincinnati, ron. Med. 64 (3), 167–177.
OH, USA. Bowler, R.M., Gocheva, V., Harris, M., et al., 2011. Neurotoxicology
ACGIH, 2013. 2013 TLVs and BEIs. ACGIH, Cincinnati, OH, USA. 32, 596–605.
Adkins Jr., B., Luginbuhl, G.H., Miller, F.J., et al., 1980. Environ. Res. Bowler, R.M., Harris, M., Gocheva, V., et al., 2012. Int. J. Hyg. Envi-
23, 110–120. ron Health 215, 393–405.
Ahmadi, F.A., Grammatopoulos, T.N., Poczobutt, A.M., et al., 2008. Bowman, A.B., Kwakye, G.F., Herrero Hernández, E., et al., 2011.
Neurochem. Res. 33, 886–901. J. Trace Elem. Med. Biol. 25 (4), 191–203.
Ahn, J., Yoo, C.I., Lee, C.R., et al., 2003. Neurotoxicology 24, 835–838. Bu, S.Y., Choi, M.K., 2012. Clin. Nutr. Res. 1 (1), 30–36.
Alessio, L., Apostoli, P., Ferioli, A., et al., 1989. Biol. Trace Elem. Res. Buchet, J.P., Lauwerys, R., Roels, H., 1976. Clin. Chim. Acta 73, 481–486.
21, 249–253. Buhmann, C., Binkofski, F., Klein, C., et al., 2005. Brain 128,
Anderson, J.G., Cooney, P.T., Erikson, K.M., 2007. Toxicol. Sci. 95, 2281–2290.
188–195. Butterworth, R.F., 2000. J. Hepatol. 32 (Suppl. 1), 171–180.
Andersen, M.E., Gearhart, J.M., Clewell, H.J., 1999. Neurotoxicology Butterworth, R.F., 2010. Neurotox. Res. 18, 100–105.
20, 161–171. Butterworth, R.F., Spahr, L., Fontaine, S., et al., 1995. Metab. Brain
Archibald, F.S., Tyree, C., 1987. Arch. Biochem. Biophys. 256, 638–650. Dis. 10, 259–267.
Arnaud, J., Favier, A., 1995. Sci. Total Environ. 159, 9–15. Calabresi, P., Ammassari-Teule, M., Gubellini, P., et al., 2001. Neuro-
Arora, M., Bradman, A., Austin, C., et al., 2012. Environ. Sci. Technol. biol. Dis. 8, 419–432.
46 (9), 5118–5125. Cahill, O.F., Bercegeay, M.S., Haggerty, R.C., et al., 1980. Toxicol.
Aschner, J.L., Aschner, M., 2005. Mol. Aspects Med. 26, 353–362. Appl. Pharmacol. 53, 83–91.
Aschner, M., 2006a. Neurotoxicology 27, 311–314. Calne, D.B., Chu, N.S., Huang, C.C., et al., 1994. Neurology 44,
Aschner, M., 2006b. Environ. Health Perspect. 114, A458. 1583–1586.
Aschner, M., 1999. Environ. Res. Section A80, 105–109. Carl, F.G., Blackwell, L.K., Barnett, F.C., et al., 1993. Epilepsia 34,
Aschner, M., Aschner, J.L., 1991. Neurosci. Biobehav. Rev. 5, 333–340. 441–446.
Aschner, M., Gannon, M., 1994. Brain Res. Bull. 33, 345–349. CEN (European Committee for Standardization), 1993. Standard
Aschner, M., Erikson, K.M., Herrero Hernández, E., 2009. Neuromo- EN481. Belgium, Brussels.
lecular Med. 11 (4), 252–266. Chandra, S.V., Ara, R., Nagar, N., et al., 1973. Acta. Biol. Med. Ger.
ATSDR (Agency for Toxic Substances and Disease Registry), 2012. 30, 857–862.
Toxicological profile for Mn. U.S. Dpt. Health Human Services, Chang, Y., Kim, Y., Woo, S.T., et al., 2009a. Neurotoxicology 30,
Publ Health Service, Agency for Toxic Substances and Disease 555–563.
Registry, Atlanta, Georgia. Chang, Y., Woo, S.T., Lee, J.J., et al., 2009b. Neurotoxicology 30,
Baek, S.Y., Cho, J.H., Kim, E.S., et al., 2004. Ind. Health 42, 315–320. 950–957.
Baldwin, M., Mergler, D., Larribe, F., et al., 1999. Neurotoxicology Chang, Y., Song, H.J., Lee, J.J., et al., 2010a. Occup. Environ. Med.
20, 343–353. 67, 809–815.
1006 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

Chang, Y., Lee, J.J., Seo, J.H., et al., 2010b. Neuroimage 53, 1279–1285. Eells, J.B., Brown, T., 2009. Neurotoxicol. Teratol. 31, 98–103.
Chen, P., Chen, Z., Li, A., et al., 2008. Biomed. Environ. Sci. 21, 233–238. Ejima, A., Imamura, T., Nakamura, S., et al., 1992. Lancet 339, 426.
Chia, S.E., Foo, S.C., Gan, S.L., et al., 1993. Scand. J. Work Environ. Elder, A., Gelein, R., Silva, V., et al., 2006. Environ. Health Perspect.
Health 19, 264–270. 114, 1172–1178.
Chia, S.E., Gan, S.L., Chua, L.H., et al., 1995. Neurotoxicology 16, Elias, Z., Mur, J.M., Pierre, F., et al., 1989. J. Occup. Med. 31, 477–483.
519–526. Ellingsen, D.G., Hetland, S.M., Thomassen, Y., 2003a. J. Environ.
Choi, Y., Park, J.K., Park, N.H., et al., 2005. J. Occup. Health 47, 68–73. Monit. 5, 84–90.
Chu, N.S., Hochberg, F.H., Calne, D.B., et al., 1995. In: Chang, L., Ellingsen, D.G., Haug, E., Ulvik, R.J., et al., 2003b. J. Appl. Toxicol.
Dyer, R., (Eds.), Marcel Dekker Inc., New York, pp. 91–103. 23 (4), 239–247.
Claus Henn, B., Ettinger, A.S., Schwartz, J., et al., 2010. Epidemiology Ellingsen, D.G., Dubeikovskaya, L., Dahl, K., et al., 2006. J. Environ.
21 (4), 433–439. Monit. 8 (10), 1078–1086.
Claus Henn, B., Schnaas, L., Ettinger, A.S., et al., 2012. Environ. Ellingsen, D.G., Chashchin, V., Haug, E., et al., 2007. Biomarkers 12
Health Perspect. 120 (1), 126–131. (5), 497–509.
Clayton, C.A., Perritt, R.L., Pellizzari, E.D., et al., 1993. J. Expo. Anal. Emara, A.M., el-Ghawabi, S.H., Madkour, O.I., et al., 1971. Br. J. Ind.
Environ. Epidemiol. 3 (no. 2), 227–250. Med. 28, 78–82.
Collipp, P.J., Chen, S.Y., Maitinsky, S., et al., 1983. Ann. Nutr. Metab. EPA (U.S. Environmental Protection Agency), 1978. Federal Reg. 43,
27, 488–494. 41424–41429.
Colomina, M.T., Domingo, J.L., Llobet, J., et al., 1996. Vet. Hum. Toxi- EPA, 1996. Integrated Risk Information System (IRIS). Carcinogenic-
col. 38, 7–9. ity assessment for lifetime exposure to Mn. US Environmental
Cooper, W.C., 1984. J. Toxicol. Environ. Health 14, 23–46. Protection Agency, Cincinnati, OH.
Couper, J., 1837. Br. Ann. Med. Pharmacol. 1, 41–42. EPA, 1993. Drinking water criteria document for Mn. Environmental
Criswell, S.R., Perlmutter, J.S., Videen, T.O., 2011. Neurology 76 (15), Protection Agency, Office of Health and Environmental Assess-
1296–1301. ment, Cincinnati, OH.
Criswell, S.R., Perlmutter, J.S., Crippin, J.S., 2012a. Arch. Neurol. 69 EPA, 1994. Reevaluation of inhalation health risks associated with
(3), 394–397. methylcyclopentadienyl Mn tricarbonyl (MMT) in gasoline. U.S.
Criswell, S.R., Perlmutter, J.S., Huang, J.L., 2012b. Occup. Environ. Environmental Protection Agency, Office of Research and Devel-
Med. 69 (6), 437–443. opment, Cincinnati, OH. EPA 600/R-94/062.
Crossgrove, J.S., Yokel, R.A., 2005. Neurotoxicology 26, 297–307. EPA, 1995. U.S. Environmental Protection Agency. Federal Reg. 60,
Crossgrove, J.S., Allen, D.D., Bukaveckas, B.L., et al., 2003. Neuro- 36414.
toxicology 24, 3–13. EPA, 2003. Health Effects Support Document for Manganese. EPA
Dastur, D.K., Manghani, D.K., Raghavendran, K.V., 1971. J. Clin. 822-R-03–003. U.S. Environmental Protection Agency, Washing-
Invest. 50, 9–20. ton, DC.
Davidsson, L., Cederblad, A., Hagebø, E., et al., 1988. J. Nutr. 118, EPA, 2007. U.S. Environmental Protection Agency. EPA454R08001.
1517–1521. http://www.epa.gov/ttnamti1/files/ambient/airtox/2006_uat
Davidsson, L., Cederblad, A., Lönnerdal, B., et al., 1989. Am. J. Clin. mp_final_report.pdf.
Nutr. 49, 170–179. EPA, 2010. U.S. Environmental Protection Agency. Technology
Davis, C.D., Greger, J.L., 1992. Am. J. Clin. Nutr. 55, 747–752. Transfer Network Air Toxics Web Site. www.epa.gov/ttn/atw/
Davis, C.D., Wolf, T.L., Greger, J.L., 1992. J. Nutr. 122, 1300–1308. hlthef/manganes.html.
Davis, C.D., Zech, L., Greger, J.L., 1993. Proc. Soc. Exp. Biol. Med. EPA, 2012a. U.S. Environmental Protection Agency. Integrated Risk
202, 103–108. Information System. http://www.epa.gov/iris/subst/0373.htm.
Davis, J.M., 1999. Neurotoxicology 20, 511–516. (accessed 27.01.14).
Davis, J.M., Malecki, E.A., Greger, J.L., 1992. Am. J. Clin. Nutr. 56, EPA, 2012b. U.S. Environmental Protection Agency. EPA 822-S-
926–932. 12-001 Office of Water. U.S. Environmental Protection Agency,
Defazio, G., Soleo, L., Zefferino, R., et al., 1996. Brain Res. Bull. 40, Washington, DC.
257–262. Ericson, J.E., Crinella, F.M., Clarke-Stewart, K.A., et al., 2007. Neuro-
Devenyi, A.G., Barron, T.F., Mamourian, A.C., 1994. Gastroenterol- toxicol. Teratol. 29, 181–187.
ogy 106, 1068–1071. Erikson, K., Aschner, M., 2002. Neurotoxicology 23, 595–602.
Dieter, H.H., Retard, W., Simon, J., et al., 1992. Die Nahrung 36, Erikson, K.M., Aschner, M., 2003. Neurochem. Int. 43, 475–480.
477–484. Erikson, K.M., Shihabi, Z.K., Aschner, J.L., et al., 2002a. Biol. Trace
Discalzi, G., Pira, E., Herrero Hernandez, E., et al., 2000. Neurotoxi- Elem. Res. 87, 143–156.
cology 21, 863–866. Erikson, K.M., Suber, R.L., Aschner, M., 2002b. Neurotoxicology 23,
Dorman, D.C., Struve, M.F., Gross, E.A., et al., 2005. Respir. Res. 6, 281–288.
121–131. Eriksson, H., Morath, C., Heilbronn, E., 1984. Acta Neurol. Scand.
Dorman, D.C., Struve, M.F., Wong, B.A., et al., 2006. Toxicol. Sci. 92, Suppl. 100, 89–93.
219–227. Eriksson, H., Gillberg, P.G., Aquilonius, S.M., et al., 1992a. Arch.
Dorman, D.C., Andersen, M.E., Roper, J.M., et al., 2012. J. Toxicol. Toxicol. 66, 359–364.
2012, 946742. Eriksson, H., Tedroff, J., Thuomas, K.A., et al., 1992b. Arch. Toxicol.
Dorner, K., Dziadzka, S., Höhn, A., et al., 1989. Br. J. Nutr. 61, 559–572. 66, 403–407.
Drown, D.B., Oberg, S.G., Sharma, R.P., 1986. J. Toxicol. Environ. Fechter, L.D., 1999. Neurotoxicology 20, 197–201.
Health 17, 201–212. Feldman, R.G., Ratner, M.H., 1999. Curr. Opin. Neurol. 12, 725–731.
Dydak, U., Jiang, Y.M., Long, L.L., et al., 2011. Environ. Health Per- Fell, J.M., Reynolds, A.P., Meadows, N., et al., 1996. Lancet 347,
spect. 119, 219–224. 1218–1221.
Eastman, R.R., Jursa, T.P., Benedetti, C., et al., 2013. Environ. Sci. Ferri, R., Donna, F., Smith, D.R., et al., 2012. J. Environ. Protect. 3,
Technol. 47 (3), 1629–1637. 374–385.
45 Manganese 1007

Finkelstein, M.M., Jerrett, M., 2007. Environ. Res. 104 (3), 420–432. Gunier, R.B., Bradman, A., Jerrett, M., et al., 2013. Environ. Sci. Tech-
Finkelstein, Y., Milatovic, D., Aschner, M., 2007. Neurotoxicology 28, nol. 47 (19), 11249–11257.
1003–1014. Gunshin, H., Mackenzie, B., Berger, U.V., et al., 1997. Nature 388,
Finley, J.W., Caton, J.S., Zhou, Z., et al., 1997. J. Nutr. 127 (12), 482–488.
2334–2341. Gunter, T.E., Pfeiffer, D.R., 1990. Am. J. Physiol. 258, C755–C786.
Fisher, A.B., Škreb, Y., 1980. Zentralbl. Bacteriol. Microbiol. Hyg. (B) Gwiazda, R., Lucchini, R., Smith, D., 2007. J. Toxicol. Environ. Health
841, 526–537. A 70, 594–605.
Fitsanakis, V.A., Au, C., Erikson, K.M., et al., 2006. Neurochem. Int. Hafeman, D., Factor-Litvak, P., Cheng, Z., et al., 2007. Environ.
48, 426–433. Health Persp. 115, 1107–1112.
Fitsanakis, V.A., Piccola, G., Marreilha dos Santos, A.P., et al., 2007. Halatek, T., Hermans, C., Broeckaert, F., et al., 1998. Eur. Respir.
Hum. Exp. Toxicol. 26, 295–302. J. 11, 726–733.
Fleming, M.D., Romano, M.A., Su, M.A., et al., 1998. Proc. Natl. Halatek, T., Sinczuk-Walczak, H., Szymczak, M., et al., 2005. Int.
Acad. Sci. U. S. A. 95, 1148–1153. J. Occup. Med. Environ. Health 18, 265–274.
Florence, T.M., Stauber, J.L., 1989. Sci. Total Environ. 78, 233–240. Halls, J.D., Fell, S.G., 1981. Anal. Chim. Acta 129, 205–211.
Flynn, M.R., Susi, P., 2010. J. Occup. Environ. Hyg. 7, 115–126. Hanzlik, R.P., Stitt, R., Traiger, G.J., 1980. Toxic. Appl. Pharmacol. 56,
FNB/IOM (U.S. Food and Nutrition Board/Institute of Medicine), 353–360.
2001. Manganese. DC. National Academy Press, Washington. Hauser, R.A., Zesiewicz, T.A., Rosemurgy, A.S., et al., 1994. Ann.
394–419. Neurol. 36, 871–875.
Forbes, J.R., Gros, P., 2003. Blood 102, 1884–1892. Hauser, R.A., Zesiewicz, T.A., Martinez, C., et al., 1996. Can. J. Neu-
Fored, C.M., Fryzek, J.P., Brandt, L., 2006. Occup. Environ. Med. 63 rol. Sci. 23, 95–98.
(2), 135–140. Haynes, E.N., Ryan, P., Chen, A., 2012. Sci. Tot. Environ. 427-428,
Fosbraey, P., Wetherell, J.R., French, M.C., 1990. J. Neurochem. 54, 19–25.
72–79. He, L., Girijashanker, K., Dalton, T.P., et al., 2006. Mol. Pharmacol.
Friedman, B.J., Freeland-Graves, J.H., Bales, C.W., et al., 1987. J. Nutr. 70, 171–180.
117, 133–143. Health Canada (2010) ISBN: 978-1-100-15221-9, Cat. No.: H128-1/
Frisbie, S.H., Mitchell, E.J., Dustin, H., et al., 2012. Environ. Health 10-600E, HC Pub.: 100122
Perspect. 120 (6), 775–778. Herndon, E.M., Jin, L., Brantley, S.L., 2011. Environ. Sci. Techn01 45
Fujishiro, H., Doi, M., Enomoto, S., et al., 2011. Metallomics 3, (1), 241–247.
710–718. Herrero Hernandez, E., Discalzi, G., Valentini, C., et al., 2006. Neuro-
Gallez, B., Baudelet, C., Adline, J., et al., 1997. Chem. Res. Toxicol. toxicology 27, 333–339.
10, 360–363. Hobbesland, A., Kjuus, H., Thelle, D.S., 1997. Scand. J. Work Envi-
Gallez, B., Baudelet, C., Adline, J., et al., 2001. Neurotoxicology 22, ron. Health 23, 334–341.
387–392. Hoet, P., Jacquerye, C., Deumer, G., et al., 2013. Clin. Chem. Lab.
Garcia, S.J., Gellein, K., Syversen, T., et al., 2007. Toxicol. Sci. 95, Med. 51 (4), 839–849.
205–214. Holbrook Jr, D.J., Washington, M.E., Leake, H.B., et al., 1975. Envi-
Garrison, A.W., et al., 1995. Environ. Toxicol. Chem. 14, 1859–1864. ron. Health Perspect. 10, 95–101.
Gavin, C.E., Gunter, K.K., Gunter, T.E., 1990. Biochem. J. 266, Hrustić, O., Šarić, M., 1980. Arh. Hig. Rada Toksikol. 31, 149–164
329–334. (Croatian with English summary).
Gavin, C.E., Gunter, K.K., Gunter, T.E., 1992. Toxicol. Appl. Pharma- HSDB (Hazardous Substances Data Bank), 2012. National Institutes
col. 115, 1–5. of Health, National Library of Medicine, Bethesda, MD. TOXNET:
Geivanidis, S., Pistikopoulos, P., Samaras, Z., 2003. Sci. Total Envi- Toxicology data network. http://toxnet.nlm.nih.gov/cgi-bin/
ron. 305, 129–141. sis/htmlgen?HSDB (accessed 27.01.14).
Gennart, J.P., Buchet, J.P., Roels, H., et al., 1992. Am. J. Epidemiol. Huang, C.C., Chu, N.S., Lu, C.S., et al., 1989. Arch. Neurol. 46,
135, 1208–1219. 1104–1106.
Gerber, G.B., Leonard, A., Hantson, P.L., 2002. Crit. Rev. Oncol. Huang, C.C., Chu, N.S., Lu, C.S., et al., 1998. Neurology 50,
Hematol. 42, 25–34. 698–700.
Gianutsos, G., Seltzer, M.D., Saymeh, R., et al., 1985. Arch. Toxicol. Huang, C.C., Weng, Y.H., Lu, C.S., et al., 2003. J. Neurol. 250,
57, 272–275. 1335–1339.
Gibbons, R.A., Dixon, S.N., Hallis, K., et al., 1976. Biochim. Biophys. Huang, C.X., Cao, C.G., 2003. J. Henan. Univ. Sci. Technol. Med. Sci.
Acta 444, 1–10. 21, 278–279.
Gil, F., Hernández, A.F., Márquez, C., et al., 2011. Sci. Total Environ. Hustvedt, S.O., Grant, D., Southon, T.E., et al., 1997. Acta Radiolog-
409, 1172–1180. ica. 38, 690–699.
Gitler, A.D., Chesi, A., Geddie, M.L., et al., 2009. Nat. Genet. 41, IMnI (International Manganese Institute), 2012. Annual Review
308–315. 2012. http://www.manganese.org (accessed 27.01.14).
Grant, D., Blazak, W.F., Brown, G.L., 1997. Acta Radiol. 38, 759–769. Inside Fuels, 2004. Inside Fuels and Vehicles, vol. 3, Inside Washing-
Graumann, R., Paris, I., Martinez-Alvarado, P., et al., 2002. Pol. J. ton Publishers, Washington, DC. no. 13.
Pharmacol. 54, 573–579. Iregren, A., 1990. Neurotoxicol. Teratol. 12, 673–675.
Greger, J.L., 1999. Neurotoxicology 20, 205–212. Iregren, A., 1999. Neurotoxicology 20, 315–324.
Guilarte, T.R., 2010. Environ. Health Persp. 118, 1071–1080. Jablonickà, A., Poláková, H., Karelová, J., et al., 1989. Mutat. Res. 224,
Guilarte, T.R., 2013. Front Aging Neurosci. 5, 23. 143–146.
Guilarte, T.R., McGlothan, J.L., Degaonkar, M., et al., 2006. Toxicol. Jacobs, R.M., Jones, A.O., Fry Jr., B.E., et al., 1978. J. Nutr. 108, 901–910.
Sci. 94, 351–358. Jang, B.-C., 2009. Toxicol. Vitro 23, 120–126.
Guilarte, T.R., Burton, N.C., McGlothan, J.L., et al., 2008. J. Neuro- Järvisalo, J., Olkinuora, M., Kiilunen, M., et al., 1992. Int. Arch.
chem. 107 (5), 1236–1247. Occup. Environ. Health 63, 495–501.
1008 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

Jiang, Y., 1996a. Chi. J. Ind. Hyg. Occup. Dis. 14, 271–273 (Chinese Kurttio, P., Vartiainen, T., Savolainen, K., 1990. Br. J. Ind. Med. 47,
with English summary). 203–206.
Jiang, Y., 1996b. Ind. Health Occup. Dis. 22, 341–343 (Chinese with Ky, S.Q., Deng, H.S., Xie, P.Y., et al., 1992. Br. J. Ind. Med. 49, 66–69.
English summary). Lai, J.C., Leung, T.K., Lim, L., 1984. Neurotoxicology 5, 37–47.
Jiang, Y.M., Mo, X.A., Du, F.Q., et al., 2006. J. Occup. Environ. Med. Lai, J.C., Chan, A.W., Leung, T.K., et al., 1992. Neurochem. Res. 17,
48, 644–649. 841–847.
Jiang, Y.M., Zheng, W., Long, L., et al., 2007. Neurotoxicology 28, Lang, C.J., 2000. Neurotoxicology 21 (N0.5), 847–855.
126–135. Laohaudomchok, W., Lin, X., Herrick, R.F., et al., 2011. J. Occup.
Jiao, J., Qi, Y., Fu, J., et al., 2008. Environ. Toxicol. Pharmacol. 26, Environ. Med. 53 (5), 506–510.
123–127. Lauwerys, R.R., Roels, H., Genet, P., et al., 1985. Am. J. Ind. Med. 7,
Johnston, C.G., Kipphut, G.W., 1988. Appl. Environ. Microbiol. 54, 171–176.
1440–1445. Le Couteur, D.G., Muller, M., Yang, M.C., et al., 2002. Rev. Environ.
Johnson, P.E., Lykken, G.J., Koryntra, E.D., 1991. J. Nutr. 121, 711–717. Health 17, 51–64.
Kafritsa, Y., Fell, J., Long, S., et al., 1998. Arch. Dis. Child. 79, 263–265. Lee, B.K., Kim, Y., 2012. Neurotoxicology 33, 401–405.
Kannurpatti, S.S., Joshi, P.G., Joshi, N.B., 2000. Neurochem. Res. 25, Lee, P.L., Gelbart, T., West, C., et al., 1998. Blood Cells Mol. Dis. 24,
1527–1536. 199–215.
Katsuragi, T., Takahashi, T.,T., Shibuya, K., et al., 1996. Rinsho Liccione, J.J., Maines, M.D., 1988. J. Pharmacol. Exp. Ther. 247, 156–161.
Shinkeigaku. 36, 780–782 (Japanese). Lim, K.O., Stark, D.D., Leese, P.T., et al., 1991. Radiology 178, 79–82.
Kawamura, R., 1941. Kitasato. Arch. Exp. Med. 18, 145–171. Lima, P.D.L., Vasconcellos, M.C., Bahia, M.O., et al., 2008. Toxicol.
Kelly, J.P., 2011. Drug Test Anal. 3 (7-8), 439–453. Vitro 22, 1032–1037.
Kern, C.H., Stanwood, G.D., Smith, D.R., 2010. Synapse 64 (5), 363–378. Lin, F.Y., 2002. Chin. J. Public Health Manage. 18, 87.
Khan, K., Wasserman, G.A., Liu, X., et al., 2012. Neurotoxicology 33 Ljung, K., Vahter, M., 2007. Environ. Health Perspect. 115, 1533–1538.
(1), 91–97. Lloyd, R.V., 1995. Chem. Res. Toxicol. 8, 111–116.
Kim, E.A., Cheong, H.K., Choi, D.S., et al., 2007. Neurotoxicology Lockman, P.R., Roder, K.E., Allen, D.D., 2001. J. Neurochem. 79,
28, 276–283. 588–594.
Kim, J.M., Kim, J.S., Jeong, S.H., et al., 2010. Neurotoxicology. 31, Lönnerdal, B., 1987. In: Kies, C. (Ed.), Nutritional biovailability of
351–355. Mn: ACS Symposium. American Chemical Society, Washington,
Kim, J.W., Kim, Y., Cheong, H.K., et al., 1998. J. Korean. Med. Sci. 13 DC, pp. 9–20. Series 354.
(N0.4), 437–439. Lönnerdal, B., 1997. Physiol. Rev. 77, 643–669.
Kim, Y., 2004. Ind. Health 42, 111–115. Loranger, S., Zayed, J., 1997. J. Air Waste Manag. Assos. 47, 983–989.
Kim, Y., 2006. Neurotoxicology 27, 369–372. Lu, L., Zhang, L.L., Li, G.J., et al., 2005. Neurotoxicology 26,
Kim, Y., 2011. In: Dushanova, J. (Ed.), Diagnostics and Rehabilita- 257–265.
tion of Parkinson’s Disease. In Tech—Open Access Publisher, Lucaciu, C.M., Dragu, C., Copăescu, L., et al., 1997. Biochim. Bio-
pp. 131–138. phys. Acta 1328, 90–98.
Kim, Y., Lee, B.-K., 2011. Neurotoxicology 32, 247–254. Lucchini, R., Zimmerman, N., 2009. Neurotoxicology 30 (6),
Kim, Y., Kim, K.S., Yang, J.S., et al., 1999a. Neurotoxicology 20, 1144–1148.
901–907. Lucchini, R., Selis, L., Folli, D., et al., 1995. Scand. J. Work Environ.
Kim, Y., Kim, J.W., Ito, K., et al., 1999b. Neurotoxicology 20, 249–252. Health 21, 143–149.
Kim, Y., Kim, J.M., Kim, J.W., et al., 2002. Mov. Disord. 17, 568–575. Lucchini, R., Bergamaschi, E., Smargiassi, A., et al., 1997. Environ.
Kim, Y., Park, J.K., Choi, Y., et al., 2005. Neurotoxicology 26, 107–111. Res. 73, 175–180.
Kim, Y., Jeong, K.S., Song, H.J., et al., 2011a. Neurotoxicology 32, Lucchini, R., Apostoli, P., Perrone, C., et al., 1999. Neurotoxicology
100–109. 20, 287–297.
Kim, Y., Bowler, R.M., Abdelouahab, N., et al., 2011b. Neurotoxicol- Lucchini, R., Albini, E., Placidi, D., et al., 2000. Neurotoxicology 21,
ogy 32 (5), 606–614. 769–775.
Kimura, M., Yagi, N., Itokawa, Y., 1978. J. Environ. Pathol. Toxicol. Lucchini, R., Albini, E., Benedetti, L., et al., 2007. Am. J. Ind. Med. 50
2, 455–461. (11), 788–800.
Kita, H., Narita, K., Van der Kloot, W., 1981. Brain Res. 205, 111–121. Lucchini, R.G., and Kim, Y., 2009. In: Vojtisek, M., Prakash, R. (Eds.),
Knopfel, M., Zhao, L., Garrick, M.D., 2005. Biochem. Mosc. 44, 3454– pp. 119–147.
3465. Lucchini, R.G., Martin, C.J., Doney, B.C., 2009. Neuromolecular Med.
Koller, W.C., Lyons, K.E., Truly, W., 2004. Neurology 62 (5), 730–733. 11 (4), 311–321.
Komura, J., Sakamoto, M., 1991. Bull. Environ. Contam. Toxicol. 46, Lucchini, R.G., Guazzetti, S., Zoni, S., et al., 2012. Neurotoxicology
921–928. 33 (4), 687–696.
Kondakis, X.G., Makris, N., Leotsinidis, M., et al., 1989. Arch. Envi- Lutz, T.A., Schroff, A., Scharrer, E., 1993. Biol. Trace Elem. 39,
ron. Health 44, 175–178. 221–227.
Korczynski, R.E., 2000. Appl. Occup. Environ. Hyg. 15, 936–945. Lyden, A., Larsson, B., Lindquist, N.G., 1984. Acta Pharmacol. Toxi-
Kostial, K., 1989. Toxicol. Ind. Health 5, 584–598. col. 55, 133–138.
Kostial, K., Blanuša, M., Piasek, M., 2005. J. Appl. Toxicol. 25, 89–93. Lytle, C.M., McKinnon, C.Z., Smith, B.N., 1994. Naturwissenschaften
Krieger, D., Krieger, S., Jansen, O., et al., 1995. Lancet 346, 270–274. 81, 509–510.
Kristensson, K., Eriksson, H., Lundh, B., et al., 1986. Acta Pharmacol. Lytle, C.M., Smith, B.N., McKinnon, C.Z., 1995. Sci. Total Environ.
Toxicol. 59, 345–348. 162, 105–109.
Kucera, J., Bencko, V., Pápayová, A., et al., 2001. Cent. Eur. J. Public. Maci, R., Arias, E., 1987. Ecotoxicol. Environ. Saf. 13, 169–173.
Health 9, 171–175. Maigetter, R.Z., Ehrlich, R., Fenters, J.D., et al., 1976. Environ. Res.
Kurttio, P., Savolainen, K., 1990. Scand. J. Work Environ. Health 16, 11, 386–391.
203–207. Malecki, E.A., 2001. Brain Res. Bull. 55, 225–228.
45 Manganese 1009

Malecki, E.A., Radzanowski, G.M., Radzanowski, T.J., et al., 1996. Newland, M.C., Cox, C., Hamada, R., et al., 1987. Fundam. Appl.
J. Nutr. 126, 489–498. Toxicol. 9, 314–328.
Mark, D., Vincent, J.H., 1986. Ann. Occup. Hyg. 30, 89–102. Newland, M.C., Ceckler, T.L., Kordower, J.H., et al., 1989. Exp. Neu-
Marreilha Dos Santos, A.P., Lopes Santos, M., Batoréu, M.C., et al., rol. 106, 251–258.
2011. Brain Res. 1382, 282–290. Nilsson, O.G., Leanza, G., Bjorklund, A., 1992. Brain Res. 584,
Martin, Ch., J., 2006. Neurotoxicology 27, 347–349. 132–140.
Mashhadi Abar Boojar, M., Goodarzi, F., 2002. J. Occup. Environ. NIOSH (National Institute for Occupational Safety and Health),
Med. 43, 282–290. 2005. Pocket guide to chemical hazards. National Institute for
McDougall, S.A., Reichel, C.M., Farley, C.M., et al., 2008. Neurosci- Occupational Safety and Health, Washington, DC.
ence 154, 848–860. Nkwenkeu, S.F., Kennedy, G., Philippe, S., et al., 2002. Sci. Total En-
McGinley, P.A., Morris, J.B., Clay, R.J., et al., 1987. Toxicol. Lett. 36, viron. 15 287 (1-2), 147–153.
137–145. Normandin, L., Kennedy, G., Zayed, J., 1999. Sci. Total Environ. 239,
Meltzer, H.M., Brantsaeter, A.L., Borch-Iohnsen, B., et al., 2010. Envi- 165–171.
ron. Res. 110, 497–504. NRC (National Research Council), 1989. Recommended dietary
Mena, I., Court, J., Fuenzalida, S., et al., 1970. New Engl. J. Med. 282, allowances. National Research Council, Washington, DC,
5–10. pp. 231–235.
Menezes-Filho, J.A., Novaes Cde, O., Moreira, J.C., et al., 2011. Envi- Oberdorster, G., and Cherian, G., 1988. In: Clarkson, T.W., Friberg, L.,
ron. Res. 111 (1), 156–163. Nordberg, G.F., Sager, P.R. (Eds.), Plenum Press, New York and
Menezes-Filho, J.A., de Carvalho-Vivas, C.F., Viana, G.F., et al., London, pp. 283–301.
2013. Neurotoxicol. doi:pii: S0161–813X(13)00154-X. 10.1016/ Oikawa, S., Hirosawa, I., Tada-Oikawa, S., et al., 2006. Free Radic.
j.neur0.2013.09.006. Biol. Med. 41, 748–756.
Méranger, J.C., Smith, D.C., 1972. Can. J. Public Health 63, 53–57. Ono, J., Harada, K., Kodaka, R., 1995. J. Parent. Enter. Nutr. 19,
Mergler, D., Baldwin, M., 1997. Environ. Res. 73, 92–100. 310–312.
Mergler, D., Huel, G., Bowler, R., et al., 1994. Environ. Res. 64, 151–180. Ordoñez-Librado, J.L., Anaya-Martinez, V., Gutierrez-Valdez, A.L.,
Mergler, D., Baldwin, M., Belanger, S., 1999. Neurotoxicology 20 (2-3), et al., 2010. Neurosci. Lett. 471 (2), 79–82.
327–342. OSHA (Occupational Safety and Health Administration), 1998.
Meyer-Baron, M., Knapp, G., Schäper, M., et al., 2009. Neurotoxicol- 29CFR 1910.1000, Occupational Safety and Health Administra-
ogy 30, 487–496. tion. Washington DC.
Meyer-Baron, M., Schäper, M., Knapp, G., et al., 2011. Toxicol. Lett. Padovani, B., Lecesne, R., Raffaelli, C., 1996. Eur. J. Radiol. 23,
206 (2), 144–151. 205–211.
Meyer-Baron, M., Schäper, M., Knapp, G., 2013. Neurotoxicology 36, Pal, P.K., Samii, A., Calne, D.B., 1999. Neurotoxicology 20, 227–238.
1–9. Park, N.H., Park, J.K., Choi, Y., et al., 2003. Neurotoxicology 24,
Miller, K.B., Caton, J.S., Finley, J.W., 2006. BioFactors 28, 33–46. 909–915.
Miller, S.T., Cotzias, G.C., Evert, H.A., 1975. Am. J. Physiol. 229, Park, J., Yoo, C.I., Sim, C.S., et al., 2006. Neurotoxicology 27 (3), 445–449.
1080–1084. Pejovic-Milic, A., Aslam, Chettle, D.R., et al., 2009. Am. J. Ind. Med.
Miller, W.J., Neathery, M.W., Gentry, R.P., et al., 1972. J. Anim. Sci. 52, 742–750.
34, 460–464. Pellizzari, E.D., Clayton, C.A., Rodes, C.E., et al., 1999. Atmospheric
Milne, D.B., Sims, R.L., Ralston, N.V., 1990. Clin. Chem. 36, 450–452. Environ. 33, 721–734.
Minoia, C., Sabbioni, E., Apostoli, P., et al., 1990. Sci. Total Environ. Pellizzari, E.D., Clayton, C.A., Rodes, C.E., et al., 2001. J. Exp. Anal.
55, 89–105. Environ. Epidemiol. 11, 423–440.
Mirowitz, S.A., Westrich, T.J., Hirsch, J.D., 1991. Radiology 181, Peneder, T.M., Scholze, P., Berger, M.L., et al., 2011. Neuroscience
117–120. 180, 280–292.
Montes, S., Schilmann, A., Riojas-Rodriguez, H., et al., 2011. Environ. Pennington, J.A.T., Young, B.E., Wilson, D.B., et al., 1986. J. Am. Diet.
Res. 111, 1302–1308. Assoc. 86, 876–891.
Morello, M., Canini, A., Mattioli, P., et al., 2008. Neurotoxicology 29, Pesch, B., Weiss, T., Kendzia, B., et al., 2012. J. Exp. Sci. Environ. Epi-
60–72. demiol. 22, 291–298.
Moreno, J.A., Streifel, K.M., Sullivan, K.A., et al., 2009. Toxicol. Sci. Pine, M., Lee, B., Dearth, R., et al., 2005. Toxicol. Sci. 85, 880–885.
112, 405–415. Pomier-Layrargues, G., Rose, C., Spahr, L., et al., 1998. Metabol.
Myers, J.E., teWaterNaude, J., Fourie, M., et al., 2003a. Neurotoxicol- Brain. Dis. 13, 311–317.
ogy 24, 649–656. Prestifilippo, J.P., Fernández-Solari, J., De Laurentiis, A., et al., 2008.
Myers, J.E., Thompson, M.L., Naik, I., et al., 2003b. Neurotoxicolgy Toxicol. Sci. 105, 295–302.
24, 875–883. Prohaska, J.R., 1987. Physiol. Rev. 67, 858–901.
Nachtman, J.P., Tubben, R.E., Commissaris, R.L., 1986. Neurobehav. Pujol, A., Pujol, J., Graus, F., et al., 1993. Neurology 43, 65–69.
Toxicol. Teratol. 8, 711–715. Racette, B.A., 2013. Neurotoxicology. http://dx.doi.org/10.1016/
Nagatomo, S., Umehara, F., Hanada, K., et al., 1999. J. Neurol. Sci. j.neur0.2013.09.007.
162, 102–105. Racette, B.A., McGee-Minnich, L., Moerlein, S.M., et al., 2001. Neu-
NAS (National Academy of Sciences), 1980. Drinking water and health, rology 56, 8–13.
vol. 3. National Academy Press, Washington, DC, pp. 331–337. Racette, B.A., Antenor, J.A., McGee-Minnich, L., et al., 2005a. Mov.
NAS/NRC (NAS National Research Council), 1989. Biologic mark- Disord. 20 (4), 492–496.
ers in reproductive toxicology. National Academy Press, Wash- Racette, B.A., Tabbal, S.D., Jennings, D., et al., 2005b. Neurology 64,
ington, DC, pp. 15–35. 230–235.
Nelson, K., Golnick, J., Korn, T., et al., 1993. Br. J. Ind. Med. 50, 510–513. Racette, B.A., Criswell, S.R., Lundin, J.I., et al., 2012. Neurotoxicol-
Newland, M.C., Weiss, B., 1992. Toxicol. Appl. Pharmacol. 113, 87–97. ogy 33 (5), 1356–1361.
1010 Roberto G. Lucchini, Michael Aschner, Yangho Kim, and Marko Šarić

Rehnberg, G.L., Hein, J.F., Carter, S.D., et al., 1980. J. Toxicol. Environ. Seibyl, J.P., Marek, K.L., Quinlan, D., et al., 1995. Ann. Neurol. 38,
Health 6, 217–226. 589–598.
Rehnberg, G.L., Hein, J.F., Carter, S.D., et al., 1982. J. Toxicol. Environ. Seth, P.K., Chandra, S.V., 1984. Neurotoxicology 5, 67–76.
Health 9, 175–188. Shen, X.M., Dryhurst, G., 1998. Chem. Res. Toxicol. 11, 824–837.
Rehnberg, G.L., Hein, J.F., Carter, S.D., et al., 1985. J. Toxicol. Environ. Shinotoh, H., et al., 1995. Neurology 45, 1199–1204.
Health 16, 887–899. Shinotoh, H., et al., 1997. Neurology 48, 1053–1056.
Reichel, C.M., Wacan, J.J., Farley, C.M., et al., 2006. Neurotoxicol. Shiotsuka, R. N., 1984. Fort Detrick, MD, US Army Medical Research
Teratol. 28, 323–332. and Development Command, (Brookhaven National Lab Report
Rentschler, G., Covolo, L., Haddad, A.A., et al., 2012. Neurotoxicol- NoBNL 35334).
ogy 33 (4), 697–702. Shukla, G.S., Chandra, V.S., 1981. Arch. Toxicol. 47, 191–196.
Riccio, A., Mattei, C., Kelsell, R.E., et al., 2002. J. Biol. Chem. 277, Sierra, P., Loranger, S., Kennedy, G., et al., 1995. Am. Ind. Hyg. Assoc.
12302–12309. J. 56, 713–716.
Rice, D.C., Lincoln, R., Martha, J., et al., 2010. J. Expo. Sci. Env. Epi- Sikk, K., Haldre, S., Aquilonius, S.M., et al., 2011. Parkinsons Dis.
demiol. 20, 634–643. 2011, 865319.
Riojas-Rodriguez, H., Solís-Vivanco, R., Schilmann, A., et al., 2010. Škreb, Y., Račić, J., Horš, N., 1980. Mutat. Res. 74, 241–242.
Environ. Health Perspect. 118 (10), 1465–1470. Smargiassi, A., Mutti, A., 1999. Neurotoxicology 20, 401–406.
Rivera-Mancía, S., Ríos, C., Montes, S., 2011. Biometals 24 (5), 811–825. Smargiassi, A., Takser, L., Masse, A., et al., 2002. Sci. Total Environ.
Robertson, G.S., Hubert, G.W., Tham, C.S., et al., 1992. Eur. J. Phar- 290, 157–164. Erratum (2002) Sci. Total Environ., 300, 247.
macol. 219, 323–325. Smith, D., Gwiazda, R., Bowler, R., et al., 2007. Am. J. Ind. Med. 50
Rodriguez-Agudelo, Y., Riojas-Rodriguez, H., Rios, C., 2006. Sci. (11), 801–811.
Total Environ. 368 (2-3), 542–556. Smith, R.A., Latchney, L.R., Senior, A.E., 1985. Biochem. Mosc 24,
Roels, H., Lauwerys, R., Genet, P., et al., 1987a. Am. J. Ind. Med. 11, 4490–4494.
297–305. Smith, R.A., Alexander, R.B., Wolman, M.G., 1987. Science 235, 1607–
Roels, H., Lauwerys, R., Buchet, J.P., et al., 1987b. Am. J. Ind. Med. 1615.
11, 307–327. Spadoni, F., Stefani, A., Morello, M., et al., 2000. Exp. Brain Res. 135,
Roels, H., Meiers, G., Delos, M., et al., 1997. Arch. Toxicol. 71, 544–551.
223–230. Spahr, L., Butterworth, R.F., Fontaine, S., et al., 1996. Hepatology 24,
Roels, H.A., Ghyselen, P., Buchet, J.P., et al., 1992. Br. J. Ind. Med. 49, 1116–1120.
25–34. Sriram, K., Lin, G.X., Jefferson, A.M., et al., 2012. Toxicology 291 (1-3),
Roels, H.A., Ortega Eslava, M.I., Ceulemans, E., et al., 1999. Neuro- 73–82.
toxicology 20, 255–271. Stamelou, M., Tuschl, K., Chong, W.K., et al., 2012. Mov. Disord. 27
Roels, H.A., Bowler, R.M., Kim, Y., et al., 2012. Neurotoxicology 33, (10), 1317–1322.
872–880. Stauber, J.L., Florence, T.M., Webster, W.S., 1987. Neurotoxicology 8,
Rose, C., Butterworth, R.F., Zayed, J., et al., 1999. Gastroenterology 431–435.
117, 640–644. Stokes, A.H., Hastings, T.G., Vrana, K.E., 1999. J. Neurosci. Res. 55,
Rosen, Y., Lenkinski, R.E., 2007. Neurotherapeutics 4, 330–345. 659–665.
Roth, J.A., 2009. Neuromolecular Med. 11 (4), 281–296. Struve, M.F., McManus, B.E., Wong, B.A., et al., 2007. Am. J. Ind.
Rukgauer, M., Klein, J., Kruse-Jarres, J.D., 1997. J. Trace. Elem. Med. Med. 50, 772–778.
Biol. 11, 92–98. Sturaro, A., Parvoli, G., Doretti, L., et al., 1994. Biol. Trace Elem. Res.
Salehi, F., Krewski, D., Mergler, D., et al., 2003. Toxicol. Appl. Phar- 40, 1–8.
macol. 191, 264–271. Sumino, K., Hayakawa, K., Shibata, T., et al., 1975. Arch. Environ.
Sakurai, H., Nishida, M., Yoshimura, T., et al., 1985. Biochim. Bio- Health 30, 487–494.
phys. Acta 841, 208–214. Suzuki, Y., Fujii, N., Yano, H., et al., 1978. Tokushima J. Exp. Med.
Sandstrom, B., Nishida, M., Yoshimura, T., et al., 1986. Acta Pharma- 25, 119–125.
col. Toxicol. 59, 60–62. Suzuki, H., Wada, O., 1981. Environ. Res. 26, 521–528.
Santos-Burgoa, C., Rios, C., Mercado, L.A., et al., 2001. Environ. Res. Takser, L., Lafond, J., Bouchard, M., et al., 2004. Environ. Res. 95,
Section A 85, 90–104. 119–125.
Šarić, M., Hrustić, O., 1975. Environ. Res. 10, 314–318. Tanner, C.M., 1992. Occup. Med. 7, 503–513.
Šarić, M., Lučić-Palaić, S., 1977. Inhaled Particles IV. Pergamon Press Tatum, V.L., Ray, A.E., Rovell-Rixx, D.C., 2001. Appl. Occup. Envi-
Ltd., Oxford, pp. 773–779. ron. Hyg. 16, 763–769.
Šarić, M., Piasek, M., 2000. Rev. Environ. Health 15 (4), 413–419. Taylor, M.D., Clewell 3rd, H.J., Andersen, M.E., et al., 2012. J. Toxicol.
Šarić, M., Markičević, A., Hrustić, O., 1977. Br. J. Ind. Med. 34, 114–118. 2012, 791431.
Šarić, M., Holetić, A., Offner, E., 1978. Proceedings of the Interna- Thomson, A.B., Olatunbosun, D., Valverg, L.S., 1971. J. Lab. Clin.
tional Conference on Heavy Metals in the Environment, vol. III. Med. 78, 642–655.
Toronto, Canada, pp. 389–398. Tjälve, H., Henriksson, J., Tallkvist, J., et al., 1996. Pharmacol. Toxi-
Sato, K., Ueda, H., Okumura, F., et al., 1994. Brain Res. 655, 233–236. col. 79, 347–356.
Schelenz, R., 1977. Radioanal. Chem. 37. 593–548. Toft, K.G., Früsk, G.A., Skotland, T., 1997. J. Pharm. Biomed. Anal.
Scholte, H.R., 1988. J. Bioenerg. Biomembr. 20, 161–191. 15, 973–981.
Schroeter, J.D., Dorman, D.C., Yoon, M., et al., 2012. Toxicol. Sci. 129 Tuschl, K., Clayton, P.T., Gospe Jr., S.M., et al., 2012. Am. J. Hum.
(2), 432–446. Genet. 90 (3), 457–466.
SCOEL (EU Scientific Committee on Occupational Exposure Limits), Verity, M.A., 1999. Neurotoxicology 20, 489–497.
2011. Recommendation from the Scientific Committee on Occu- Vigeh, M., Yokoyama, K., Ramezanzadeh, F., et al., 2008. Reprod.
pational Exposure Limits for manganese and inorganic manga- Toxicol. 25, 219–223.
nese compounds. Eur. Comm. Employment Soc. Aff. Inclusion Vitarella, D., Wong, B.A., Moss, O.R., et al., 2000. Toxicol. Appl. Phar-
Health Saf. work. SCOEL/Sum/127, June 2011. macol. 163, 279–285.
45 Manganese 1011

Walker, R.C., Purnell, G.L., Jones-Jackson, L.B., et al., 2004. Neuro- Wilson, D.C., Tubman, R., Bell, N., et al., 1991. Early Hum. Dev. 26,
toxicology 25, 533–542. 223–226.
Walton, A.P., Wei, G.T., Liang, Z., et al., 1991. Anal. Chem. 63, 232–240. Wirth, J.J., Rossano, M.G., Daly, D.C., et al., 2007. Epidemiology 18,
Wang, C., Gordon, P.B., Hustvedt, S.O., et al., 1997. Acta Radiologica. 270–273.
38, 665–676. Wittczak, T., Dudek, W., Krakowiak, A., et al., 2008. Int. J. Occup.
Wang, D.X., Du, X.Q., Zheng, W., 2008. Toxicol. Lett. 176, 40–47. Med. Environ. Health 21, 81–83.
Wang, S.H., Wu, J., 2000. Prevent. Med. 25, 145–146. Wolters, E.C., Huang, C.C., Clark, C., et al., 1989. Ann. Neurol. 26,
Wasserman, G.A., Liu, X., Parvez, F., et al., 2006. Environ. Health Per- 647–651.
spect. 114 (1), 124–129. Wongwit, W., Kaewkungwal, J., Chantachum, Y., et al., 2004. South-
Wassermann, M., Mihail, G., 1961. Arch. Gewerbepath. Gewerbe- east Asian J. Trop. Med. Public Health 35, 764–769.
hyg. 18, 632–657. Wright, R.O., Amarasiriwardena, C., Woolf, A.D., et al., 2006. Neuro-
Wedekind, K.J., Titgemeyer, E.C., Twardock, A.R., et al., 1991. J. Nutr. toxicol. 27 (2), 210–216.
121, 1776–1786. Wu, W., Zhang, Y., Zhang, P., et al., 1996. Chin. J. Prev. Med. 30,
Weiss, B., 2006. Neurotoxicology 27 (3), 362–368. 266–268.
Weissman, N., 1981. Klin. Wochenschr. 59, 413–421. Yamada, M., Ohno, S., Okayasu, I., et al., 1986. Acta Neuropatolog-
Welder, F. C., 1994. In: Klimis-Tavantzis, D. J. (Ed.), CRC Press, Boca ica. 70, 273–278.
Raton, LA, pp. 1–36. Yang, H., Sun, Y., Zheng, X., 2007. J. Biochem. Mol. Toxicol. 21, 94–100.
Wennberg, A., Iregren, A., Struwe, G., et al., 1991. Scand. J. Work Yiin, S.J., Lin, T.H., Shih, T.S., 1996. Scand. J. Work. Environ. Health
Environ. Health 17, 255–262. 22, 381–386.
WHO (World Health Organization), 1981. Manganese. World Health Yin, Z., Jiang, H., Lee, E.S., et al., 2010. J. Neurochem. 112, 1190–1198.
Organization. International Programme on Chemical Safety (En- Yokel, R.A., 2009. Neuromolecular Med. 11 (4), 297–310.
vironmental Health Criteria 17), Geneva, Switzerland. Yokel, R.A., Crossgrove, J.S., 2004. Res. Rep. Health Eff. Inst. 119,
WHO, 1986. Diseases caused by Mn and its toxic compounds. Early 7–58.
detection of occupational diseases. World Health Organization, Zatta, P., Lucchini, R., van Rensburg, S.J., et al., 2003. Brain Res. Bull.
Geneva, Switzerland. 62, 15–28.
WHO, 1994. Concise International Chemical Assessment Document Zayed, J., Vyskocil, A., Kennedy, G., 1999. Int. Arch. Occup. Environ.
63 Manganese And Its Compounds: Environmental Aspects. Health 72, 7–13.
ISBN 92 4 153063 4. Zerón, H.M., Rodríguez, M.R., Montes, S., et al., 2011. J. Trace Elem.
WHO, 2000. Manganese. Air Quality Guidelines, second ed. World Med. Biol. 25 (4), 225–229.
Health Organization, Regional Office for Europe, Copenhagen, Zhang, S., Fu, J., Zhou, Z., 2004. Toxicol. Vitro 18, 71–77.
Denmark (Chapter 6.8). pp. 154–156. Zheng, W., Fu, S.X., Dydak, U., et al., 2011. Neurotoxicology 32, 1–8.
WHO, 2004a. Manganese and its compounds : environmental as- Zielhuis, R.L., del Castilho, P., Herber, R.F., et al., 1978. Environ.
pects. ISBN 92 4 153063 4. World Health Organization, Geneva, Health Perspect. 25, 103–109.
Switzerland. Zoni, S., Albini, E., Lucchini, R., 2007. Am. J. Ind. Med. 50 (11),
WHO, 2004b. third ed. Guidelines for Drinking-Water Quality, 812–830.
vol 1, Recommendations. World Health Organization, Geneva: Zoni, S., Bonetti, G., Lucchini, R.G., 2012. J. Trace Elem. Med. Biol.
Switzerland. 26 (2-3), 179–182.
WHO, 2011. Guidelines for drinking-water quality, fourth ed. World Zoratti, M., Szabo, I., 1995. Biochim. Biophys. Acta 1241, 139–176.
Health Organization, Geneva, Switzerland. ISBN 978 92 4 154815 Zota, A.R., Ettinger, A.S., Bouchard, M., et al., 2009. Epidemiology
1WHO Press. 20, 367–373.

You might also like