Você está na página 1de 10

Environmental Toxicology and Pharmacology 60 (2018) 100–109

Contents lists available at ScienceDirect

Environmental Toxicology and Pharmacology


journal homepage: www.elsevier.com/locate/etap

Elevated heavy metals levels in cognitively impaired patients from Pakistan T


a b b a,⁎
Ghazala Iqbal , Wahid Zada , Abdul Mannan , Touqeer Ahmed
a
Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology,
Sector H-12, Islamabad, 44000, Pakistan
b
Department of Pharmacy, COMSATS Institute of Information Technology, Abbottabad, 22060, K.P.K, Pakistan

A R T I C LE I N FO A B S T R A C T

Keywords: The deficit in the visuospatial skills, thinking, learning and memory is termed as cognitive impairment. Human
Copper exposure to heavy metals is a potential risk factor for developing cognitive impairment. This study aimed to
Lead investigate the possible association between the concentration of heavy metals and the extent of cognitive im-
Aluminum pairment. Blood samples were subjected to microwave assisted acid digestion and evaluated for metals levels
Zinc
using atomic absorption spectrophotometry. We measured the levels of Cu, Pb, Al, Zn, Cd and Mn in 183 patients
Cadmium
diagnosed with cognitive impairment; mild (n = 72), moderate (n = 86) and severe (n = 25) and compared
Manganese
Oxidative stress them to age-matched healthy controls (n = 90). Results showed that all the aforementioned elements were
Mini mental state examination significantly higher in cognitively impaired patients and increasing concentration was strongly correlated with
Age increase in severity of the disease. The correlation study has shown that among the studied metals, Al and Cu are
Metal accumulation strongly associated with the cognitive impairment. This suggests the need for decrease in metal exposure to
humans from environment, food and industries.

1. Introduction uptake mechanism exits to provide essential trace element (such as Zn


and Cu) to the body (Clemens, 2006). This transport mechanism is used
Cognition impairment is the diminished ability of an individual to by all the heavy metals to enter and get accumulated in the body. These
know the world (Folstein et al., 1985). Cognitive impairment is a accumulated heavy metals have the capacity to generate reactive
condition in which an individual’s cognitive functions such as re- oxygen species (superoxide and nitric oxide) in biological system and
membering, language, attention and visuospatial abilities are impaired lead to oxidative stress. This results in increased production of reactive
and affect their everyday life (Giagulli et al., 2016). This leads to de- oxygen species which overwhelms body antioxidant potential and in-
creased activity and deficit in learning and memory; consequently in- duces various proteins modification and DNA damage, resulting in
creased risk of disability and health care cost (Plassman et al., 2008). various diseases including neurological disorders (Jomova and Valko,
The cognitive abilities are influenced by various factors, such as; ge- 2011). Oxidative stress impairs the function of endothelial cells and
netics, environment, diet, aging and life style (Deary et al., 2009). The promotes penetration of macrophages to parenchyma of brain, conse-
deficit in cognitive functions are increasing globally and it is predicted quently the nutrients availability to brain cells are decreased and in-
to increase proportionately more in developing countries (Mathers itiate the inflammatory process (Markesbery, 1997). Studies have
et al., 2008). During the process of aging, there is a decline in health shown that elevated levels of metals are associated with various dis-
and cognitive functions, which is usually proportionally related to the orders enlisted in Table 1.
increased level of oxidative stress and abnormality in protein ag- In Pakistan, we are exposed to heavy metals mainly from environ-
gregation (Mattson and Magnus, 2006). Along with this if the in- mental and dietary sources as mentioned in Table 1. A relevant study by
dividuals are exposed to various toxins which are responsible for Azizullah et al., has shown very high levels of heavy metals (Zn, Cu,
causing oxidative stress, the susceptibility of worsening the cognitive Mn, Cd, Pb, Ni and Hg) in drinking water throughout in the Pakistan.
functions is significantly increased (Meramat et al., 2017). These levels are significantly higher than those set by WHO (Azizullah
The exposure of humans to heavy metals from occupation and en- et al., 2011). Textile, pharmaceuticals and fertilizer industries are the
vironment is a potential risk factor for cognitive impairment (Afridi main source of metal contamination in water of Pakistan (Nasrullah
et al., 2011). The metals cannot be synthesized or destroyed by the et al., 2006). The crops irrigated with metal (Cu, Zn, Mn. Pb, Co and Ni)
human body. Therefore, an efficient absorption, transport and cellular contaminated water is another main source of human exposure to


Corresponding author.
E-mail address: touqeer.ahmed@asab.nust.edu.pk (T. Ahmed).

https://doi.org/10.1016/j.etap.2018.04.011
Received 16 January 2018; Received in revised form 10 April 2018; Accepted 11 April 2018
Available online 16 April 2018
1382-6689/ © 2018 Elsevier B.V. All rights reserved.
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

heavy metals in Pakistan (Hussain et al., 2013). Hence, these metals


and the oxidative stress produced by these metals are the main culprits
Safe intake quantities of
metal in drinking water
(Organization, 2008) for various diseases including cognitive impairment in developing
countries (Olness, 2003), like Pakistan.
Studies have revealed that elevated levels of metals are associated
with dementia, neurotoxicity, amyloid plaques formation, and

0.005 ppm
0.05 ppm
0.1 ppm

0.4 ppm
Alzheimer’s disease (mentioned in Table 1). However, there is very
1 ppm

5 ppm
limited data available on metal exposure and its effects on cognitive
impairment, especially, from Pakistan where people are highly exposed
Permissible limits of metals in

to metals. Therefore this study was designed to evaluate the effect of


body (Organization, 2008;

metal accumulation with severity of cognitive impairment (measured


through MMSE scores).

0.26 mg/l; 0.06 mg/kg


0.1 mg/l; 0.5 μg/kg

The determination of elements in the human tissue, is the most


Singh et al., 2011)

0.1 mg/l; 25 μg/kg

0.06 mg/l; 7 μg/kg


15 mg/l; 1 mg/kg

common and important application for clinical screening and diag-


nostic procedures. Blood sample may be conveniently used to study
0.1 μg/kg

bio-indicator for this purpose (Kazi et al., 2008c). The evaluation of


trace elements in blood samples is frequently used traditional ap-
proach, however any change in physiological or environmental con-
ditions may result in fluctuation of these elements in the blood (Afridi
Parkinson’s disease (Hirsch et al., 1991), dementia

Motor neuron disease such as Amyotrophic lateral


Alzheimer’s disease (Cuajungco and Fagét, 2003).
Motor neuron diseases (Weihl and Lopate, 2006),

Alzheimer’s disease (Crapper McLachlan, 1986),

and Schneider, 2003; Needleman and Gatsonis,


attention deficit hyperactivity disorder (Lidsky
Neurotoxicity, risk of mild mental retardation,

et al., 2008).
Based on toxicity of these metals and limited data showing the re-
Parkinson’s disease (Powers et al., 2003),
Alzheimer’s disease (Syme et al., 2004).

lationship of these metals with cognitive impairment, we decided to


neurotoxicity (Mergler et al., 1998).

accurately measure the trace element concentration in “blood samples”


Nervous system related disorders

and to measure their correlation with “cognitive impairment”.


sclerosis (Bar-Sela et al., 2001).
1990), lead encephalopathy.

Therefore it demands a highly sensitive technique. Atomic absorption


spectrometry is the extensively used method due to its selectivity and
(Larson et al., 1992).

sensitivity, and numerous reports justify this statement (Afridi et al.,


2011; Kazi et al., 2008a,c). The primary goal of studies was to measure
the metal concentration in blood samples and find out if association
exits with cognitive impairment. Moreover, we also evaluated if metal
accumulation was also related with age or not. It is the first study to
document the association between the concentrations of metals in
whole blood with the age in Pakistani subjects.
Anemia (Lozoff et al., 2000), diabetes mellitus (Kazi

metabolism, a possible higher risk of kidney stones


Kidney damage, disturbed phosphorus and calcium

(Nordberg, 2004), bone damage (Kazantzis, 1979),


Developmental disabilities (Lidsky and Schneider,
Cancer, cardiovascular disease (Kok et al., 1988),
et al., 1991), cancer, cardiovascular disease (Kok
Diseases associated with elevated level of metals

Bone disorder (Baxter et al., 1953), hepatitis (Li

pulmonary dysfunction (Mazzoli-Rocha et al.,


Ischemic heart disease (Costello et al., 2014),

2. Materials and methods


chronic kidney disease (Kurella et al., 2004),

2.1. Compliance with ethical standards

All procedures performed in this study, involving human partici-


cancer (Waalkes et al., 1988).

pants, were in accordance with ethical standard of Internal Review


Board (IRB); letter number IRB-67, Atta-ur-Rahman School of Applied
Biosciences, National University of Sciences and Technology and with
diabetes mellitus.

the 1964 Helsinki declaration. Informed consent was obtained from all
Disease associated with elevated level of metals and their permissible limits.

et al., 2008b).
et al., 1988).

individual participants included in the study.


2010).

2003)

2.2. Assessment of cognitive function

Neuropsychiatric evaluation of all subjects was performed by heath


water treatment plants, antacids (Keith et al., 2008)

Contaminated food, drinking-water, the lead-glazed

Color pigment in paint, in the fabrication of nickel-

products anticorrosive agent, a neutron-absorber in


Diet, drinking water (Bouchard et al., 2011), metal
Beverages cans and foil, antiperspirants, cosmetics,

nuclear power plants, tobacco smoking(Orlowski

care provider. The inclusions criteria of subject are listed in Table 2. All
Food (Galdes and Vallee, 1983), zinc fumes, soil

material for food storage, lead-containing paint,

cadmium batteries, fertilizers, stabilizer in PVC

273 subjects were interviewed to obtain detailed information through


laden dust in industrial areas (Finley, 2004).

an approved questionnaire that covers the demographic data, medical


history and occupation. The cognitive function of each subject was
assessed using mini mental state examination (MMSE). The MMSE also
Fish, meat (Wong et al., 2001)

known as Folstein test, is a 30-point questionnaire test that is used


(Fine et al., 1997), Refineries.

extensively in research and clinical setting to screen cognitive impair-


ment (Folstein et al., 1975). All subjects were then classified into mild,
and Piotrowski, 2003).
Environmental sources

moderate and severe cognitive impairment, according to their MMSE


score as listed in Table 3.

2.3. Measurement of metal levels


petrol.

The analysis of heavy metals was carried out by means of a double-


beam Perkin Elmer atomic absorption spectrometer model 700 (Perkin
Manganese
Aluminum

Cadmium

Elmer, USA) equipped with a flame burner and graphite furnace, a


Copper
Table 1

Metal

Lead
Zinc

pyrocoated graphite tube with an integrated platform and an auto-


sampler AS-800 (Perkin Elmer). The instrumental parameters are

101
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

shown in Table 4. The stock standard solution was prepared according Table 3
to instructions provided in manual of the instrument. The straight line Classification of subjects according to MMSE score.
was obtained while plotting the absorbance of standard vs. concentra- Groups MMSE Score(Crum et al., Number of
tion. Different standard dilutions were used for each metals and the 1993; Sayed et al., 2014) subjects
absorbance of test samples were only measured after getting a straight
Age-matched healthy Control 25–30 90
line graph. If the concentration in test sample was high, then it was
Mild Cognitive Impairment 21–24 72
further diluted. The standard dilutions for Zn were 0.5 mg/L, 1 mg/L, (Mild CI)
1.5 mg/L; Pb were 2 mg/L, 4 mg/L, 6 mg/L; Cu, Al, Mn and Cd were Moderate Cognitive Impairment 10–20 86
1 mg/L, 2 mg/L, 3 mg/L. A KENTAX (KNM-20/BW) domestic micro- (Moderate CI)
wave oven (maximum heating power of 700 W) was used for digestion Severe Cognitive Impairment 0–9 25
(Severe CI)
of the biological samples.

2.4. Reagents and glasswares using “Graph Pad Prism” software and taken as significant only if the p
value was less than 0.05. One-way ANOVA was applied using
Concentrated nitric acid (65%) and hydrogen peroxide (30%) were BonferroniPost-hoc test. Correlation analysis of “metal concentration”
obtained from Merck (Darmstadt, Germany). Working standard solu- with “MMSE scores” and “metal concentration” with “age” was per-
tions of Al, Cu, Pb, Cd, Mn and Zn were prepared prior to their use by formed using Pearson’s correlation test using Graph Pad Prism.
stepwise dilution of certified standard solutions. Before use, all glass-
ware and plastic materials were soaked for 24 h in 5 M nitric acid,
washed and finally rinsed with pure water and dried. Double distilled 3. Results
and syringe filtered (0.2 μm) water was used throughout the work.
3.1. Demographic data
2.5. Blood sample collection and pretreatment
The demographic data obtained from all the groups i.e. age-matched
Before the sample collection, all healthy and cognitively impaired healthy control, mild CI, moderate CI and severe CI is summarized in
patients were informed about the study and all agreed to participate. Table 5. Among the studied groups the percentage of female was high in
Venous blood samples (3 mL) were collected in EDTA K3 vacutainer severe cognitively impaired group. It was 60% in case of severe CI,
tubes (Bio Tube, Inc.), and the whole blood sample was thoroughly 59.30% in moderate CI, 54.20% in mild CI and 46.6% in age-matched
mixed and transferred to a storage boxes at −4 °C until further pro- healthy controls; while the percentage of male was 40%, 40.70%,
cessed for acid digestion. 45.8% and 53.4% respectively. The income status is categorized into 3
groups i.e. lower, middle and upper according to the dollars earned per
2.6. Microwave-assisted acid digestion of blood samples for metal detection person per day (Durr-e-Nayab, 2011). Lower income status subjects
were; severe CI 64%, Moderate CI 53%, Mild CI 25% and age-matched
For digestion of blood samples, 0.5 ml of heparinized human whole healthy control 12.5%; in case of middle income status these were 32%,
blood samples of each patient and control subjects were directly taken 24.4%, 57%, 30% respectively; and in case of upper class income status
into separate, properly labeled test tube. The mixture of concentrated these were 4%, 22.6%, 18%, 57.3% respectively.
HNO3–H2O2 (2:1, v/v) was freshly prepared and three milliliters of this The educations level is categorized according to the years of edu-
mixture was added in each test tube, thoroughly mixed and kept at cation, in to 4 groups i.e. 0–5 years, 6–10 years. 11–14 years and 14+
room temperature for 10 min. This mixture was then heated in micro- years of education. Among the 0–5 years groups subjects were, severe
wave oven at 500 W. Microwave digestion was carried out to shorten CI 68%, moderate CI 50%, mild CI 48.6% and age-matched healthy
digestion time. Complete digestion of blood samples required 1–2 min. control 28.9%, while in case of 6-10 years group,32%, 45.3%, 44.4%
After cooling, the digested samples were filtered through Whatman 42 and 54.4%, in case of 11-14 years group 0%, 3.5%, 4.2%, 5.6% and in
filter paper and brought to a required volume with pure water (Afridi case of 14+ years, 0%, 1.2%, 2.8% and 11.1% respectively.
et al., 2011; Kazi et al., 2007, 2008b; Olmedo et al., 2010). After fil- The occupation of the mild CI, moderate CI, severe CI and age
tration, the samples were ready for the measurement of metal by the matched healthy control is mentioned in Table 6. The groups are fur-
atomic absorption spectrometer. ther divided into male and female and their occupation is mentioned.

2.7. Statistical analysis 3.2. Copper concentration in blood samples

Data were expressed as mean ± standard error of mean (SEM) and The levels of Cu in all the biological samples of mild, moderate and
n = number of subjects. Statistical analysis of results was performed severe cognitive impairment patients were higher as compared to the

Table 2
Inclusion and exclusion criteria.
Inclusion Criteria Exclusion Criteria

Objective evidence (from physical and neurological examination and laboratory tests usually Suggestive evidence for an alternative causation of the mental disorder, e.g. a
(MMSE) Mini mental state examination). highly loaded family history for a clinically similar or related disorders
The presence of a disorder in cognitive function for most of the time for at least two weeks, as Known genetic neurologic disorder or history of psychiatric illness
reported by the individual or a reliable informer
History of cerebral disease, damage or dysfunction, or of systemic physical disorder known to Systemic physical disorder
cause cerebral dysfunction, including hormonal disturbances and non-psychoactive drug
effects
Either gender Obvious kidney disease (Kidney failure)
Individuals with age ≥50 years. Subject carrier of blood transmitted infections
Irrespective of Race and origin Known liver disease (Hepatitis, jaundice)
Patients having cancer/ tumors

102
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Table 4
Atomic absorption spectrometer parameters for heavy metal analysis.
Parameters Aluminum Zinc Lead Copper Cadmium Manganese

Lamp current 10.0 mA 5.0 mA 5.0 mA 3.0 mA 4.0 mA 6.0 mA


Slit width 0.2 nm 0.2 nm 0.2 nm 0.2 nm 0.2 nm 0.2 nm
Wave length 309.3 nm 213 nm 405.8 nm 327.4 nm 228.8 nm 285.2 nm
Sensitivity 0.55 μg/ml 0.008 μg/ml 0.06 μg/ml 0.025 μg/ml 0.5 μg/ml 0.3 μg/ml

age-matched healthy control subjects. The level of Cu in severe cogni- 3.6. Manganese concentration in blood samples
tive impairment (2.37 ± 0.12 mg/L) was higher (p < 0.001) from
mild (1.41 ± 0.10 mg/L) and moderate cognitive impairment Mn levels were significantly (p < 0.001) higher in severe
(1.97 ± 0.11 mg/L). There was significantly(p < 0.001) decreased Cu (92.08 ± 6.8 μg/L) and moderate (77.8 ± 2.4 μg/L) cognitively im-
level in age-matched healthy control (0.7 ± 0.06 mg/L) relative to paired group as compared to the age-matched healthy control group
mild cognitive impairment (Fig. 1a). (52.8 ± 2.8 μg/L). The mild (64.97 ± 3.76 μg/L; p < 0.05) cogni-
tively impaired group was also having significantly elevated levels of
Mn compared to the age-matched healthy control group (Fig. 1e).
3.3. Aluminum concentration in blood samples
3.7. Cadmium concentration in blood samples
The high concentration of Al was associated with the decrease in
MMSE score. The level of Al in blood samples were significantly Cd concentration in mild (3.87 ± 0.25 μg/L; p < 0.05), moderate
(p < 0.001) higher in severe cognitive impairment subjects (4.86 ± 0.27 μg/L; p < 0.001) and severe (6.11 ± 0.38 μg/L;
(127.8 ± 4.83 μg/L) when compared to mild (63.37 ± 3.52 μg/L) and p < 0.001) cognitively impaired groups were significantly high when
moderate (92.4 ± 3.57 μg/L) cognitive impairment groups (Fig. 1b). compared to the age-matched healthy control (2.85 ± 0.19 μg/L)
Al concentration was significantly (p < 0.001) low in age-matched group (Fig. 1f).
healthy control group (47.30 ± 2.76 μg/L) when compared to cogni-
tively impaired groups.
3.8. Correlation analysis of metal levels with MMSE

3.4. Zinc concentration in blood samples In order to examine the association of different metals concentra-
tions on the extent of cognitive impairment, correlation analysis was
The concentration of Zn was significantly high in the moderate performed. Pearson’s correlation test revealed the negative correlation
(6.4 ± 0.26 mg/L; p < 0.01) and severe (7.92 ± 0.32 mg/L; between the metal concentration and MMSE score. The maximum
p < 0.001) cognitive impairment groups when compared to the age- correlation was observed with Al (r = −0.638; p < 0.001) followed by
matched healthy control group (4.95 ± 0.33 mg/L). There was no Cu (r = −0.610; p < 0.001), Pb (r = −0.554; p < 0.001), Cd
significant difference in concentration of Zn among age-matched (r = −0.418; p < 0.001), Mn (r = −0.417; p < 0.001) and Zn
healthy control and mild cognitively impaired group (5.98 ± 0.24 mg/ (r = −0.329; p < 0.001) respectively (Fig. 2a–f).
L) but the increasing trend was there (Fig. 1c).
3.9. Correlation analysis of metal levels with age

3.5. Lead concentration in blood samples Correlation analysis was done in order to examine the relationship
of different metals concentrations with the age. Pearson’s correlation
The concentration of Pb was significantly (p < 0.001) high in se- test was performed and it showed the correlation between the metal
vere (274.1 ± 11.92 μg/L) cognitively impaired group as compared to concentration and age in years of all the subjects studied in all the
the age-matched healthy control group (157.6 ± 7.76 μg/L). Moderate groups. Among the studied metals, the maximum correlation was ob-
(229.2 ± 6.93 μg/L; p < 0.001) and mild (188.7 ± 6.23 μg/L; served with Al (r = 0.187; p < 0.01) followed by Pb (r = 0.148;
p < 0.05) cognitively impaired groups had significantly higher con- p = p < 0.05) and Cd (r = 0.14; p < 0.05). There was no correlation
centration of Pb, relative to the age-matched healthy control group observed with the concentration of Cu (r = −0.025), Zn(r = 0.111)
(Fig. 1d). and Mn (r = 0.106) with the age (Fig. 3a–f).

Table 5
Demographic data of subjects included in study.
Age-matched healthy Control Mild CI Moderate CI Severe CI P-value

Percent Male 53.4 45.80 40.70 40

Percent Female 46.6 54.20 59.30 60

Age ( ± SEM) 58.1 ± 0.69 61.7 ± 1.071 65.1 ± 1.18 79.68 ± 1.4 < 0.0001

Percent Income status


Lower 12.2 25 53 64
Middle 30 57 24.4 32
Higher 57.3 18 22.6 4

Education in years (percent)


0–5 28.9 48.6 50 68
6–10 54.4 44.4 45.3 32
11–14 5.6 4.2 3.5 0
14+ 11.1 2.8 1.2 0

103
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Table 6
: Occupation of the subjects included in the study.
Groups No of Male Occupation No of Female Occupation

Age-matched healthy control 48 8 teachers; 11 Govt. servant; 27 private; business; 2 labor 42 6 Home tuitions; 14 Tailor; 22 House wife

Mild 33 12 Govt. servant; 21 private business 39 1 lady health worker; 2 maid; 36 house wife

Moderate 51 3 school teachers; 21 private business; 12 mason; 13 labor; 2 Tailor 35 4 maid; 31 house wife

Severe 10 2 driver; 2 mason; 6 labor 15 2 maid; 13 house wife

3.10. Metal levels in sub-fractions of the control group according to the age 2004; Praticò et al., 2002; Smorgon et al., 2004).There are several
evidences which highlight the role of oxidative stress mechanisms and
The average age of control subjects and the average age of mild, free radical damage in pathology of cognitive impairment (Solfrizzi
moderate and severe cognitive impairment groups was different, et al., 2006). In this study, we determine the Cu, Pb, Al, Zn, Cd and Mn
therefore, we wanted to check the effect on metal accumulation with levels in acid digested blood samples from the patients of cognitive
increasing age. The control group was sub-fractioned to 50–59 years, impairment and compared them with the age-matched healthy control
60–69 years and 70–79 years; to evaluate whether metal accumulation subjects. The degree of cognitive impairment was measured by MMSE
is related to the age or not. There was no significant difference observed score. MMSE score measures all five dimensions of cognition which are
in the metal concentration among the sub-fractions of age-matched specified by the diagnostic and statistical manual (DSM-iv) that in-
healthy control. Only the Cu concentration was decreasing with the cludes; memory impairment (both short and long term), aphasia (de-
increase in age. All the other metals showed no change (there was no terioration of language function), apraxia (impaired ability to execute
increase) as shown in Fig. 4a–f. motor activities), agnosia (inability to recognize objects) and dis-
turbances in executive functioning.
Despite the unavailability of accurate data about the prevalence of
4. Discussion cognitive impairment in Pakistan, there are various studies that in-
dicate that dementia and mental disorders are increasing in developing
This population based study explored the association between the countries like Pakistan, either due to poverty or illiteracy (Ferri et al.,
levels of metals in blood to the extent of cognitive impairment. The 2006; Patel and Kleinman, 2003; Prince et al., 2003). Cognitive im-
heavy metals exposure causes oxidative stress in brain tissue and is a pairment is classified as mild, moderate and severe cognitive
risk factor for cognitive impairment (Deibel et al., 1996; Huang et al.,

Fig. 1. Bar graph showing the metals concentration in blood samples of age-matched healthy control, mild cognitive impairment, moderate cognitive impairment
and severe cognitively impaired subjects. (a) Cu concentration in blood samples. (b) Al level in blood samples.(c) Zn level in blood samples. (d) Pb level in blood
samples. (e) Mn level in blood samples and (f) Cd blood level comparison among groups. *p < 0.05, **p < 0.01, ***p < 0.001 compared with age-matched
healthy control group; # p < 0.05, ##p < 0.01, ###p < 0.001 comparison with mild cognitive impaired group; n is sample size.

104
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Fig. 2. Correlation graph is between the metal concentration in blood of studied samples and MMSE score. (a) Correlation of Cu concentration with MMSE scores. (b)
Correlation of Al concentration with MMSE scores. (c) Correlation of Zn concentration with MMSE scores. (d) Correlation of Pb concentration with MMSE scores. (e)
Correlation of Mn concentration with MMSE scores and (f) Correlation of Cd concentration with MMSE scores.

impairment based on MMSE scores. In mild cognitive impairment, the impairment as compared to Pb, Mn, Cd and Zn. The negative correla-
learning and memory is compromised to a greater extend as can be tion between metal concentration and MMSE score depict that as the
expected at certain age. However the moderate and severe cognitive metal concentration is increasing, there is decreasing trend of MMSE
impairment is much severe condition and patient sometimes need score. The correlation analysis are supporting the literature that Al le-
nursing care (Crum et al., 1993; Sayed et al., 2014). There are various vels are strongly associated with cognitive impairment and can leads
risk factors which are associated with the cognitive impairment and towards dementia (Rondeau et al., 2000). We found that the con-
metals may be a suspect risk factor for decreased level of cognition. A centration of Cu, Zn and Mn in blood is not associated with the increase
population based survey in French men has shown that metals and pH in age. On the other hand, the correlation among the age and Al, Cd and
of drinking water has been related to cognitive impairment in elderly Pb concentration depicted that with the increase in age these metals
(Jacqmin et al., 1994). In Pakistan, heavy metals are extensively found were also increasing. But the association between metal concentration
in environment and humans are exposed to these trace metals from and age is weaker than the relationship of metal concentration with the
many sources, such as contaminated, water, air, soil and food (Jomova MMSE score. Hence, based on the strong association of metals with
and Valko, 2011). Numerous studies point out that these metals act as MMSE score, we concluding that elevated metals concentration is a risk
catalysts in the oxidative reactions at cellular level in biological func- factor for the cognitive impairment and its progression.
tions (Deibel et al., 1996; Patra et al., 2001; Valko et al., 2005). Metals Our study demonstrates that individuals with high concentration of
induced oxidative stress results in high yield of reactive oxygen species Al in blood have greater cognitive impairment. The strongest correla-
in neuronal cells and this might be responsible for the neurotoxic effects tion of Al concentration was shown with the MMSE score. Previous
of these metals (Barnham et al., 2004; Ercal et al., 2001; Iqbal et al., studies also reports that Al from environmental source leads to the
2016; Squitti et al., 2011). The oxidative stress act as an early signal to dementia and cognitive decline (Solfrizzi et al., 2006). Al has been
mediate apoptosis of neurons and cerebral endothelial cells of blood shown to convert amyloid beta alpha-helix conformation to beta-ple-
brain barrier (Bush, 2000; Greenlund et al., 1995). This damage of ated sheets thereby causing aggregation of amyloid protein and for-
blood brain barrier makes it more vulnerable to neurotoxicants (Oteiza mation of amyloid plaques(Exley et al., 1993) and neuronal death, thus
et al., 2004), further worsening the situation and leading to mental leading to cognitive impairment (Exley et al., 1993).Our study has
disorders. We found that as the concentration of these metals in blood is shown that with increase in age, the Al concentration is also increasing,
increasing, the cognitive impairment shifts from mild to moderate and and it is consistent with the previous finding reporting that Al accu-
then severe. This is strongly proven by our correlation studies between mulate in age dependent manner in aorta and cerebral artery (Minami
metal concentrations with MMSE score. Among the examined metals, Al et al., 1996). But further studies are needed for detailed elaboration and
and Cu are found to be more associated with the extent of cognitive underlying mechanism exploration.

105
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Fig. 3. Correlation graph showing the relationship between metal concentrations in blood samples of all the subjects and their age in years. (a) Correlation of Cu
concentration with age in years. (b) Correlation of Al concentration with age in years. (c) Correlation of Zn concentration with age in years. (d) Correlation of Pb
concentration with age in years. (e) Correlation of Mn concentration with age in years and (f) Correlation of Cd concentration with age in years.

In our study, Cu was at second number for its association with absorbed by respiratory tract as compared to digestive tract and smo-
MMSE. Squitti et al, have also reported that Cu concentration can help kers are more prone to Cd toxicity (Elinder et al., 1983). Cd up-reg-
to discriminate the cognitively impaired subjects from the control ulates glutamate dehydrogenase and enhances excitotoxicity of gluta-
subjects (Squitti et al., 2011). Cu act as a cofactor for cytochrome C mate as well as reduces superoxide dismutase level. All these factors
oxidase and superoxide dismutase enzymes which is involved in redox result in impairment of blood brain barrier and ultimately cognitive
reactions (Valko et al., 2005). Cu induces neurotoxicity by two me- functions (Bar-Sela et al., 2013). Our results indicate that severity of the
chanisms, first, Cu directly catalyzes the formation of reactive oxygen cognitive impairment is significantly increased as the concentration of
species (Prousek, 2007), and, secondly high level of Cu decrease glu- Cd in blood is elevated. These results are consistent with the previously
tathione level, which is a powerful antioxidant in cell. Decreased glu- reported data mentioning that the people who are exposed to Cd have
tathione level further aggravates oxidative stress in cell(Mattie and greater tendency to have poor cognitive functions (Hart et al., 1989).
Freedman, 2004). Cu accumulation in blood is independent of the age Further studies are needed to find Cd pathological effects alone and in
as documented in the results. Wills et al., found that Cu and Zn con- combination in old age population.
centration build up in retina of eye with age(Wills et al., 2008), but The values of Mn in mild, moderate and severe cognitively impaired
there is no data available about Cu accumulation in the whole blood individuals were higher as compared to age-matched healthy controls
with age. hence highlighting the toxic role of Mn in cognitive pathology. Mn
Blood Pb concentration can affect various physiological functions rapidly crosses blood brain barrier and causes neurotoxicity. Chronic
(Kim et al., 1996)and early life exposure to Pb leads to the formation of Mn exposure leads to Mn psychosis (Verhoeven et al., 2011). The use of
amyloid plaques (Wu et al., 2008). Previous data reported that there is Mn containing pesticides is widespread in agriculture, hence increasing
higher possibility of cognitive impairment in individuals who are more the human exposure to this toxic metal (Lucchini et al., 1995).Ger-
exposed to environmental Pb (Rogan et al., 2001). Our results con- hardsson et al, have previously reported that increased serum Mn level
firmed that Pb at high concentration in blood is associated with im- is directly related to problems in cognitive functions (Gerhardsson
paired cognitive functions as confirmed from correlation studies and et al., 2008).
the patients with severe cognitive impairment showed the highest blood Zn and Cu are essential elements for human health but overexposure
concentration of Pb as compared to age-matched healthy control sub- can lead to various health complications (Arora et al., 2006; Fosmire,
jects. 1990). Zn at higher concentration act as neurotoxin causing demyeli-
There is no known physiological function of Cd and it is absorbed nation (Tokuda et al., 2007), oxidative stress and leads to neuronal cell
and transported by the system acquired by other essential metals death at different concentrations (Koh et al., 1996). Previous study has
(Satarug et al., 2011) from the environment and diet. Cd is readily reported that Zn is involved in the pathogenesis of dementia (Mizuno

106
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Fig. 4. The bar graph showing the sub-fractions of the control group according to the age and metal levels. (a) Cu concentration in sub-fractions of age-matched
healthy control. (b) Al concentration in sub-fractions of age-matched healthy control. (c) Zn concentration in sub-fractions of age-matched healthy control. (d) Pb
concentration in sub-fractions of age-matched healthy control. (e) Mn concentration in sub-fractions of age-matched healthy control. (f) Cd concentration in sub-
fractions of age-matched healthy control. *p < 0.05.

and Kawahara, 2013). Our study also showed the association of the need for decrease in metal exposure to humans especially Al and Cu
high Zn concentration in serum with the level of cognitive impairment. from environment, food and industries.
There is marked increase in Zn level in individuals with low MMSE
score. Declaration of interest
The significantly high levels of all the aforementioned metals were
observed to be associated with decrease in MMSE score. Numerous The authors declare that they have no conflict of interest.
studies from Pakistan have shown the relationship of high metal con-
centration in biological samples with various diseases including pa- Acknowledgments
ralysis (Afridi et al., 2011), diabetes mellitus (Afridi et al., 2008), lung
cancer (Kazi et al., 2008a) and myocardial infarction (Kazi et al., The authors will like to thank Atta-ur-Rahman School of Applied
2008c) but limited data is present on cognitive functions. Conclusively, Biosciences, National University of Sciences and Technology, Islamabad
oxidative stress caused by these metals may be considered as culprit for and Higher Education Commission (HEC) of Pakistan for sponsoring
development and progression of cognitive dysfunctions. The persistence this project.
of metals in the environment and diet requires a long term approach to
restrict our exposure by environmental management and maintaining References
of lower toxic metals level in water and diet. Along with this the
awareness should be provided to the general public about the toxic Giagulli, A.V., Guastamacchia, E., Licchelli, B., Triggiani, V., 2016. Serum testosterone
effects of metals on nervous system. and cognitive function in ageing male: updating the evidence. Recent Pat. Endocr.
Metab. Immune Drug Discov. 10, 22–30.
Afridi, H.I., Kazi, T.G., Kazi, A.G., Shah, F., Wadhwa, S.K., Kolachi, N.F., Shah, A.Q., Baig,
5. Conclusion J.A., Kazi, N., 2011. Levels of arsenic, cadmium, lead, manganese and zinc in bio-
logical samples of paralysed steel mill workers with related to controls. Biol. Trace
Elem. Res. 144, 164–182.
This study conclusively suggests that metals exposure is associated Afridi, H.I., Kazi, T.G., Kazi, N., Jamali, M.K., Arain, M.B., Jalbani, N., Baig, J.A., Sarfraz,
with pathology of cognitive impairment and its progression in old age. R.A., 2008. Evaluation of status of toxic metals in biological samples of diabetes
mellitus patients. Diabetes Res. Clin. Pract. 80, 280–288.
There could be multiple mechanisms responsible for the cognitive im- Arora, R., Kulshreshtha, S., Mohan, G., Singh, M., Sharma, P., 2006. Estimation of serum
pairment caused by metal exposure but, the oxidative stress seems to be zinc and copper in children with acute diarrhea. Biol. Trace Elem. Res. 114, 121–126.
the most significant. However single element could not be responsible Azizullah, A., Khattak, M.N.K., Richter, P., Häder, D.-P., 2011. Water pollution in
Pakistan and its impact on public health—a review. Environ. Int. 37, 479–497.
for all various types of cellular and molecular events. This suggests the

107
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Bar-Sela, S., Reingold, S., Richter, E.D., 2001. Amyotrophic lateral sclerosis in a battery- Kazi, T., Memon, A., Afridi, H., Jamali, M., Arain, M., Jalbani, N., Sarfraz, R., 2008a.
factory worker exposed to cadmium. Int. J. Occup. Environ. Health 7, 109–112. Determination of cadmium in whole blood and scalp hair samples of Pakistani male
Bar-Sela, S., Reingold, S., Richter, E.D., 2013. Amyotrophic lateral sclerosis in a battery- lung cancer patients by electrothermal atomic absorption spectrometer. Sci. Total
factory worker exposed to cadmium. Int. J. Occup. Environ. Health 7 (2), 109–112. Environ. 389, 270–276.
Barnham, K.J., Masters, C.L., Bush, A.I., 2004. Neurodegenerative diseases and oxidative Kazi, T.G., Afridi, H.I., Jamali, M.K., Arain, M.B., Jalbani, N., Syed, N., 2007. Evaluation
stress. Nat. Rev. Drug Discov. 3, 205–214. of zinc status in whole blood and scalp hair of female cancer patients. Clin. Chim.
Baxter, J., Van Wyk, J., Follis Jr, R., 1953. A bone disorder associated with copper de- Acta 379, 66–70.
ficiency. 2. Histological and chemical studies on the bones. Bull. Johns Hopkins Hosp. Kazi, T.G., Afridi, H.I., Kazi, N., Jamali, M.K., Arain, M.B., Jalbani, N., Kandhro, G.A.,
93, 25–39. 2008b. Copper, chromium, manganese, iron, nickel, and zinc levels in biological
Bouchard, M.F., Sauvé, S., Barbeau, B., Legrand, M., Brodeur, M.-È., Bouffard, T., samples of diabetes mellitus patients. Biol. Trace Elem. Res. 122, 1–18.
Limoges, E., Bellinger, D.C., Mergler, D., 2011. Intellectual impairment in school-age Kazi, T.G., Afridi, H.I., Kazi, N., Jamali, M.K., Arain, M.B., Sarfraz, R.A., Jalbani, N.,
children exposed to manganese from drinking water. Environ. Health Perspect. 119, Ansari, R., Shah, A.Q., Khandhro, G.A., 2008c. Distribution of zinc, copper and iron in
138. biological samples of Pakistani myocardial infarction (1st, 2nd and 3rd heart attack)
Bush, A.I., 2000. Metals and neuroscience. Curr. Opin. Chem. Biol. 4, 184–191. patients and controls. Clin. Chim. Acta 389, 114–119.
Clemens, S., 2006. Toxic metal accumulation, responses to exposure and mechanisms of Keith, S., Jones, D., Rosemond, Z., Ingerman, L., Chappell, L., 2008. Toxicological Profile
tolerance in plants. Biochimie 88, 1707–1719. for Aluminum. Atlanta, GA.
Costello, S., Brown, D.M., Noth, E.M., Cantley, L., Slade, M.D., Tessier-Sherman, B., Kim, R., Rotnitzky, A., Sparrow, D., Weiss, S.T., Wager, C., Hu, H., 1996. A longitudinal
Hammond, S.K., Eisen, E.A., Cullen, M.R., 2014. Incident ischemic heart disease and study of low-level lead exposure and impairment of renal function: the normative
recent occupational exposure to particulate matter in an aluminum cohort. J. Expos. aging study. JAMA 275, 1177–1181.
Sci. Environ. Epidemiol. 24, 82–88. Koh, J.-Y., Suh, S.W., Gwag, B.J., He, Y.Y., 1996. The role of zinc in selective neuronal
Crapper McLachlan, D.R., 1986. Aluminum and Alzheimer’s disease. Neurobiol. Aging 7 death after transient global cerebral ischemia. Science 272, 1013.
(6), 525–532. Kok, F.J., Van Duijin, C.M., Hofman, A., Van Der Voet, G.B., De Wolff, F.A., Paays, C.H.C.,
Crum, R.M., Anthony, J.C., Bassett, S.S., Folstein, M.F., 1993. Population-based norms for Valkenburg, H.A., 1988. Serum copper and zinc and the risk of death from cancer and
the mini-mental state examination by age and educational level. JAMA 269, cardiovascular disease. Am. J. Epidemiol. 128, 352–359.
2386–2391. Kurella, M., Chertow, G.M., Luan, J., Yaffe, K., 2004. Cognitive impairment in chronic
Cuajungco, M.P., Fagét, K.Y., 2003. Zinc takes the center stage: its paradoxical role in kidney disease. J. Am. Geriatr. Soc. 52, 1863–1869.
Alzheimer's disease. Brain Res. Rev. 41, 44–56. Larson, E.B., Kukull, W.A., Katzman, R.L., 1992. Cognitive impairment: dementia and
Deary, I.J., Corley, J., Gow, A.J., Harris, S.E., Houlihan, L.M., Marioni, R.E., Penke, L., Alzheimer's disease. Annu. Rev. Public Health 13, 431–449.
Rafnsson, S.B., Starr, J.M., 2009. Age-associated cognitive decline. Br. Med. Bull. 92, Li, Y., Togashi, Y., Sato, S., Emoto, T., Kang, J., Takeichi, N., Kobayashi, H., Kojima, Y.,
135–152. Une, Y., Uchino, J., 1991. Spontaneous hepatic copper accumulation in Long-Evans
Deibel, M., Ehmann, W., Markesbery, W., 1996. Copper, iron, and zinc imbalances in Cinnamon rats with hereditary hepatitis. A model of Wilson’s disease. J. Clin. Invest.
severely degenerated brain regions in Alzheimer's disease: possible relation to oxi- 87, 1858.
dative stress. J. Neurol. Sci. 143, 137–142. Lidsky, T.I., Schneider, J.S., 2003. Lead neurotoxicity in children: basic mechanisms and
Durr-e-Nayab, 2011. Estimating the middle class in Pakistan. Pak. Dev. Rev. 1–28. clinical correlates. Brain 126, 5–19.
Elinder, C.G., Kjellström, T., Lind, B., Linnman, L., Piscator, M., Sundstedt, K., 1983. Lozoff, B., Jimenez, E., Hagen, J., Mollen, E., Wolf, A.W., 2000. Poorer behavioral and
Cadmium exposure from smoking cigarettes: variations with time and country where developmental outcome more than 10 years after treatment for iron deficiency in
purchased. Environ. Res. 32, 220–227. infancy. Pediatrics 105, e51.
Ercal, N., Gurer-Orhan, H., Aykin-Burns, N., 2001. Toxic metals and oxidative stress part Lucchini, R., Selis, L., Folli, D., Apostoli, P., Mutti, A., Vanoni, O., Iregren, A., Alessio, L.,
I: mechanisms involved in metal-induced oxidative damage. Curr. Top. Med. Chem. 1995. Neurobehavioral effects of manganese in workers from a ferroalloy plant after
1, 529–539. temporary cessation of exposure. Scand. J. Work. Environ. Health 143–149.
Exley, C., Price, N.C., Kelly, S.M., Birchall, J.D., 1993. An interaction of β‐amyloid with Markesbery, W.R., 1997. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic.
aluminium in vitro. FEBS Lett. 324, 293–295. Biol. Med. 23, 134–147.
Ferri, C.P., Prince, M., Brayne, C., Brodaty, H., Fratiglioni, L., Ganguli, M., Hall, K., Mathers, C., Fat, D.M., Boerma, J.T., 2008. The Global Burden of Disease: 2004 Update.
Hasegawa, K., Hendrie, H., Huang, Y., 2006. Global prevalence of dementia: a Delphi World Health Organization.
consensus study. Lancet 366, 2112–2117. Mattie, M.D., Freedman, J.H., 2004. Copper-inducible transcription: regulation by metal-
Fine, J.M., Gordon, T., Chen, L.C., Kinney, P., Falcone, G., Beckett, W.S., 1997. Metal and oxidative stressresponsive pathways. Am. J. Physiol.-Cell Physiol. 286,
fume fever: characterization of clinical and plasma IL-6 responses in controlled C293–C301.
human exposures to zinc oxide fume at and below the threshold limit value. J. Occup. Mattson, M.P., Magnus, T., 2006. Aging and neuronal vulnerability. Nat. Rev. Neurosci. 7,
Environ. Med. 39, 722–726. 278–294.
Finley, J.W., 2004. Does environmental exposure to manganese pose a health risk to Mazzoli-Rocha, F., dos Santos, A.N., Fernandes, S., Ferreira Normando, V.M., Malm, O.,
healthy adults? Nutr. Rev. 62, 148. Nascimento Saldiva, P.H., Wanderley Picanço-Diniz, D.L., Faffe, D.S., Zin, W.A.,
Folstein, M., Anthony, J.C., Parhad, I., Duffy, B., Gruenberg, E.M., 1985. The meaning of 2010. Pulmonary function and histological impairment in mice after acute exposure
cognitive impairment in the elderly. J. Am. Geriatr. Soc. 33, 228–235. to aluminum dust. Inhal. Toxicol. 22, 861–867.
Folstein, M.F., Folstein, S.E., McHugh, P.R., 1975. “Mini-mental state”: a practical method Meramat, A., Rajab, N.F., Shahar, S., Sharif, R.A., 2017. DNA damage, copper and lead
for grading the cognitive state of patients for the clinician. J. Psychiatr. Res. 12, associates with cognitive function among older adults. J. Nutr. Health Aging 21,
189–198. 539–545.
Fosmire, G.J., 1990. Zinc toxicity. Am. J. Clin. Nutr. 51, 225–227. Mergler, D., Baldwin, M., Belanger, S., Larribe, F., Beuter, A., Bowler, R., Panisset, M.,
Galdes, A., Vallee, B.L., 1983. Categories of zinc metalloenzymes. Met. Ions Biol. Syst. 15, Edwards, R., De Geoffroy, A., Sassine, M., 1998. Manganese neurotoxicity, a con-
1–54. tinuum of dysfunction: results from a community based study. Neurotoxicology 20,
Gerhardsson, L., Lundh, T., Minthon, L., Londos, E., 2008. Metal concentrations in plasma 327–342.
and cerebrospinal fluid in patients with Alzheimer’s disease. Dement. Geriatr. Cogn. Minami, T., Ichii, M., Tohno, Y., Tohno, S., Utsumi, M., Yamada, M.-o., Okazaki, Y., 1996.
Disord. 25, 508–515. Age-dependent aluminum accumulation in the human aorta and cerebral artery. Biol.
Greenlund, L.J., Deckwerth, T.L., Johnson, E.M., 1995. Superoxide dismutase delays Trace Elem. Res. 55, 199–205.
neuronal apoptosis: a role for reactive oxygen species in programmed neuronal death. Mizuno, D., Kawahara, M., 2013. The molecular mechanisms of zinc neurotoxicity and
Neuron 14, 303–315. the pathogenesis of vascular type Senile Dementia. Int. J. Mol. Sci. 14, 22067.
Hart, R.P., Rose, C.S., Hamer, R.M., 1989. Neuropsychological effects of occupational Nasrullah, R.N., Bibi, H., Iqbal, M., Durrani, M.I., 2006. Pollution load in industrial ef-
exposure to cadmium. J. Clin. Exp. Neuropsychol. 11, 933–943. fluent and ground water of Gadoon Amazai Induatrial Estate (GAIE) Swabi, NWFP. J.
Hirsch, E., Brandel, J.P., Galle, P., Javoy- Agid, F., Agid, Y., 1991. Iron and aluminum Agric. Biol. Sci. 1, 18–24.
increase in the substantia nigra of patients with Parkinson’s disease: an X‐Ray mi- Needleman, H.L., Gatsonis, C.A., 1990. Low-level lead exposure and the IQ of children: a
croanalysis. J. Neurochem. 56, 446–451. meta-analysis of modern studies. JAMA 263, 673–678.
Huang, X., Moir, R.D., Tanzi, R.E., Bush, A.I., Rogers, J.T., 2004. Redox‐Active metals, Nordberg, G.F., 2004. Cadmium and health in the 21st century–historical remarks and
oxidative stress, and Alzheimer’s disease pathology. Ann. N.Y. Acad. Sci. 1012, trends for the future. Biometals 17, 485–489.
153–163. Olmedo, P., Pla, A., Hernández, A., López-Guarnido, O., Rodrigo, L., Gil, F., 2010.
Hussain, A., Alamzeb, S., Begum, S., 2013. Accumulation of heavy metals in edible parts Validation of a method to quantify chromium, cadmium, manganese, nickel and lead
of vegetables irrigated with waste water and their daily intake to adults and children, in human whole blood, urine, saliva and hair samples by electrothermal atomic ab-
District Mardan, Pakistan. Food Chem. 136, 1515–1523. sorption spectrometry. Anal. Chim. Acta 659, 60–67.
Iqbal, G., Iqbal, A., Mahboob, A., Farhat, S.M., Ahmed, T., 2016. Memory enhancing Olness, K., 2003. Effects on brain development leading to cognitive impairment: a
effect of black pepper in the AlCl3 induced neurotoxicity mouse model is mediated worldwide epidemic. J. Dev. Behav. Pediatr. 24, 120–130.
through its active component chavicine. Curr. Pharm. Biotechnol. 17, 962–973. Organization, W.H, 2008. Guidelines for Drinking-Water Quality [Electronic Resource]:
Jacqmin, H., Commenges, D., Letenneur, L., Barberger-Gateau, P., Dartigues, J.-F., 1994. Incorporating 1st and 2nd Addenda, vol. 1 World Health Organization
Components of drinking water and risk of cognitive impairment in the elderly. Am. J. Recommendations.
Epidemiol. 139, 48–57. Orlowski, C., Piotrowski, J.K., 2003. Biological levels of cadmium and zinc in the small
Jomova, K., Valko, M., 2011. Advances in metal-induced oxidative stress and human intestine of non-occupationally exposed human subjects. Hum. Exp. Toxicol. 22.
disease. Toxicology 283, 65–87. Oteiza, P.I., Mackenzie, G.G., Verstraeten, S.V., 2004. Metals in neurodegeneration: in-
Kazantzis, G., 1979. Renal tubular dysfunction and abnormalities of calcium metabolism volvement of oxidants and oxidant-sensitive transcription factors. Mol. Aspects Med.
in cadmium workers. Environ. Health Perspect. 28, 155. 25, 103–115.

108
G. Iqbal et al. Environmental Toxicology and Pharmacology 60 (2018) 100–109

Patel, V., Kleinman, A., 2003. Poverty and common mental disorders in developing Solfrizzi, V., Colacicco, A.M., D’Introno, A., Capurso, C., Parigi, A.D., Capurso, S.A.,
countries. Bull. World Health Organ. 81, 609–615. Torres, F., Capurso, A., Panza, F., 2006. Macronutrients, aluminium from drinking
Patra, R., Swarup, D., Dwivedi, S., 2001. Antioxidant effects of α tocopherol, ascorbic acid water and foods, and other metals in cognitive decline and dementia. J. Alzheimer’s
and L-methionine on lead induced oxidative stress to the liver, kidney and brain in Dis. 10, 303–330.
rats. Toxicology 162, 81–88. Squitti, R., Ghidoni, R., Scrascia, F., Benussi, L., Panetta, V., Pasqualetti, P., Moffa, F.,
Plassman, B.L., Langa, K.M., Fisher, G.G., Heeringa, S.G., Weir, D.R., Ofstedal, M.B., Bernardini, S., Ventriglia, M., Binetti, G., 2011. Free copper distinguishes mild cog-
Burke, J.R., Hurd, M.D., Potter, G.G., Rodgers, W.L., 2008. Prevalence of cognitive nitive impairment subjects from healthy elderly individuals. J. Alzheimer’s Dis. 23,
impairment without dementia in the United States. Ann. Intern. Med. 148, 427–434. 239–248.
Powers, K., Smith-Weller, T., Franklin, G., Longstreth, W., Swanson, P., Checkoway, H., Syme, C.D., Nadal, R.C., Rigby, S.E., Viles, J.H., 2004. Copper binding to the Amyloid-β
2003. Parkinson’s disease risks associated with dietary iron, manganese, and other (Aβ) peptide associated with Alzheimer’s disease folding, coordination geometry, ph
nutrient intakes. Neurology 60, 1761–1766. dependence, stoichiometry, and affinity of Aβ-(1–28): Insights from a range of
Praticò, D., Uryu, K., Sung, S., Tang, S., Trojanowski, J.Q., Lee, V.M.-Y., 2002. Aluminum complementary spectroscopic techniques. J. Biol. Chem. 279, 18169–18177.
modulates brain amyloidosis through oxidative stress in APP transgenic mice. FASEB Tokuda, E., Ono, S.-I., Ishige, K., Naganuma, A., Ito, Y., Suzuki, T., 2007. Metallothionein
J. 16, 1138–1140. proteins expression, copper and zinc concentrations, and lipid peroxidation level in a
Prince, M., Acosta, D., Chiu, H., Scazufca, M., Varghese, M., Group, D.R., 2003. Dementia rodent model for amyotrophic lateral sclerosis. Toxicology 229, 33–41.
diagnosis in developing countries: a cross-cultural validation study. Lancet 361, Valko, M., Morris, H., Cronin, M., 2005. Metals, toxicity and oxidative stress. Curr. Med.
909–917. Chem. 12, 1161–1208.
Prousek, J., 2007. Fenton chemistry in biology and medicine. Pure Appl. Chem. 79, Verhoeven, W.M., Egger, J.I., Kuijpers, H.J., 2011. Manganese and acute paranoid psy-
2325–2338. chosis: a case report. J. Med. Case Rep. 5, 1.
Rogan, W.J., Dietrich, K.N., Ware, J.H., Dockery, D.W., Salganik, M., Radcliffe, J., Jones, Waalkes, M.P., Rehm, S., Riggs, C.W., Bare, R.M., Devor, D.E., Poirier, L.A., Wenk, M.L.,
R.L., Ragan, N.B., Chisolm Jr., J.J., Rhoads, G.G., 2001. The effect of chelation Henneman, J.R., Balaschak, M.S., 1988. Cadmium carcinogenesis in male Wistar [Crl:
therapy with succimer on neuropsychological development in children exposed to (WI) BR] rats: dose-response analysis of tumor induction in the prostate and testes
lead. N. Engl. J. Med. 344, 1421–1426. and at the injection site. Cancer Res. 48, 4656–4663.
Rondeau, V., Commenges, D., Jacqmin-Gadda, H., Dartigues, J.-F., 2000. Relation be- Weihl, C.C., Lopate, G., 2006. Motor neuron disease associated with copper deficiency.
tween aluminum concentrations in drinking water and Alzheimer's disease: an 8-year Muscle Nerve 34, 789–793.
follow-up study. Am. J. Epidemiol. 152, 59–66. Wills, N.K., Sadagopa Ramanujam, V.M., Kalariya, N., Lewis, J.R., van Kuijk, F.J.G.M.,
Satarug, S., Garrett, S.H., Sens, M.A., Sens, D.A., 2011. Cadmium, environmental ex- 2008. Copper and zinc distribution in the human retina: relationship to cadmium
posure, and health outcomes. Ciencia Saude Coletiva 16, 2587–2602. accumulation, age, and gender. Exp. Eye Res. 87, 80–88.
Sayed, K., Taha, T.T., Saad, M., Mohsen, L.A., Ibrahiem, M.A., Fadeel, N.A., Sotouhy, A., Wong, W.Y., Flik, G., Groenen, P.M.W., Swinkels, D.W., Thomas, C.M.G., Copius-
2014. Is mini‐mental score examination scoring a new predictor of uncontrolled Peereboom, J.H.J., Merkus, H.M.W.M., Steegers-Theunissen, R.P.M., 2001. The im-
hypertension? J. Clin. Hypertens. 16, 348–353. pact of calcium, magnesium, zinc, and copper in blood and seminal plasma on semen
Singh, R., Gautam, N., Mishra, A., Gupta, R., 2011. Heavy metals and living systems: an parameters in men. Reprod. Toxicol. 15, 131–136.
overview. Indian J. Pharmacol. 43, 246. Wu, J., Basha, M.R., Brock, B., Cox, D.P., Cardozo-Pelaez, F., McPherson, C.A., Harry, J.,
Smorgon, C., Mari, E., Atti, A.R., Dalla Nora, E., Zamboni, P.F., Calzoni, F., Passaro, A., Rice, D.C., Maloney, B., Chen, D., 2008. Alzheimer's disease (AD)-like pathology in
Fellin, R., 2004. TRACE ELEMENTS AND COGNITIVE IMPAIRMENT: AN ELDERLY aged monkeys after infantile exposure to environmental metal lead (Pb): evidence for
COHORT STUDY. Arch. Gerontol. Geriatr. 38, 393–402. a developmental origin and environmental link for AD. J. Neurosci. 28, 3–9.

109

Você também pode gostar