Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

C H A P T E R

16

Cardiovascular Disease
BENGT SJÖGREN, CAROLINA BIGERT, AND PER GUSTAVSSON

ABSTRACT CVD is a group of diseases that includes coronary


heart disease (CHD), cerebrovascular disease, periph-
There is strong epidemiological and mechanistic eral arterial disease, rheumatic heart disease, deep
evidence for a causal association between exposure to vein thrombosis, and pulmonary embolism (WHO,
arsenic and lead and development of cardiovascular 2012). Sometimes the term circulatory diseases is used
disease. There is relatively strong but less conclusive to encompass all of these diseases. CHD is a disease of
evidence for a causal relationship between cadmium the blood vessels supplying the heart muscle. This con-
and mercury exposure and cardiovascular disease. All dition is often called ischemic heart disease (IHD) and
of these metals are suspected of inducing pathophysi- acute myocardial infarction belongs to this entity. Cere-
ological changes relevant to atherogenic disorders, brovascular diseases are subdivided into ischemic and
including increased oxidative stress, inflammatory hemorrhagic diseases. Stroke is a widely used unspe-
response, and coagulation activity. cific term for a group of cerebrovascular diseases of
Cardiomyopathy has been associated with exces- abrupt onset that cause neurological damage. Approx-
sive intake of arsenic, cobalt, and iron. Deficiency of imately 85% of strokes are caused by inadequate blood
selenium has been related to dilated cardiomyopathy. flow to the brain, i.e. ischemic stroke. Hemorrhagic
Airborne beryllium exposure has been associated strokes are subdivided into hemorrhage into the brain
with heart disease, including both ischemic heart tissue and hemorrhage into the subarachnoid space
disease and cor pulmonale. Signs of cor pulmo- (Zivin, 2004). The main representative of peripheral
nale have also been observed after cobalt exposure arterial disease is hypertension. Terminology used in
among hard-metal workers. Pulmonary fibrosis with the different studies was retained in the following text.
increased blood pressure in the pulmonary circula- The most common cause of CVD is atherosclerosis.
tion is a likely mechanism in the development of cor Atherosclerosis has an inflammatory nature, which
pulmonale. was described by the Austrian pathologist Carl von
Ionizing irradiation may explain the suggested rela- Rokitansky in the 1840s and by Rudolf Virchow some-
tion between uranium exposure and cerebrovascular what later. Rokitansky believed that inflammation
disease. was secondary to other disease processes and Virchow
promoted atherosclerosis as a primary inflammatory
disease (Frostegård, 2010; Mayerl et al., 2006). The
1 INTRODUCTION response-to-injury hypothesis was summarized by
Ross (1993). This theory postulates alteration to the
Cardiovascular disease (CVD) is the leading cause endothelium and intima, due to for example mechani-
of death in the developed as well as in the develop- cal injury, toxins, and oxygen radicals, as the initiating
ing world. The annual mortality of CVD is expected event leading to endothelial dysfunction. During the
to reach 23.6 million by 2030 (Alissa and Ferns, 2011). last two decades more data has linked inflammation

Handbook on the Toxicology of Metals 4E


http://dx.doi.org/10.1016/B978-0-444-59453-2.00016-0 313 Copyright © 2015 Elsevier B.V. All rights reserved.
314 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

to the occurrence of atherosclerosis and thrombosis A total of 7026 Canadian aluminum primary pro-
(Epstein et al., 1999; Libby et al., 2002; Ridker, 1999; duction workers using Söderberg pots were followed
Ross, 1999). Several markers of inflammation such between 1957 and 1999 and compared with the
as interleukin-6 (IL-6), fibrinogen, C-reactive protein province’s population. Smoking-adjusted internal
­
(CRP), serum amyloid A protein, and increased leu- comparisons were conducted using Cox regression
kocyte cell count are established risk factors for IHD for the male subjects (n = 6423). Cumulative benzo[a]
(Danesh et al., 1998, 2000, 2005, 2008). pyrene exposure was associated with IHD mortality
Environmental studies focus on the general popula- (HR 1.62, 95% CI 1.06-2.46) in the highest exposure cat-
tion, and the exposure route is almost always ingestion egory (Friesen et al., 2010).
of metals or metal-containing compounds. Prospective In summary, studies of aluminum exposure and
studies are more conclusive because the exposure by CVD are scarce and consequently no firm conclusions
definition occurs prior to the effect or disease outcome. can be drawn.
Consequently, cross-sectional studies are regarded as
less conclusive. Occupational studies focus on a spe-
cific occupational group or a specific metal exposure, 3 ARSENIC
and the exposure route is inhalation as a rule. In many
studies of occupational exposures, the CVD mortal- The most common sources of exposure to arsenic
ity for one specific occupational group is compared are contaminated drinking water and occupational
with the national mortality. Such comparisons may be exposure among primary copper smelter workers.
biased because the general population includes sick CVD is one of several important outcomes of arsenic
and disabled people who are unable to work. exposure. These diseases comprise peripheral vascular
This is a condensed review, mainly focusing on epi- disease including blackfoot disease, IHD, stroke, and
demiological studies of environmental as well as occu- hypertension. These effects are mentioned in Chapter
pational exposures to metals and metal-containing 28 on arsenic.
compounds and their relations with CVD. In 1900 in the Manchester area in England an epi-
demic of a multisystem disease occurred, with over
6000 cases and more than 70 deaths. This epidemic has
2 ALUMINUM been described as the first metal-induced cardiotoxic
syndrome, and was produced by the intake of beer
Aluminum is ubiquitous in the environment and contaminated with arsenic (Alexander, 1969). The syn-
comprises about 8% of the Earth’s crust. Aluminum drome included the usual signs and symptoms of arse-
can be absorbed by ingestion or inhalation. Accumula- nic poisoning, and an unusual but prominent aspect of
tion in the human body relates to the presence of alu- this epidemic was heart failure. The source of arsenic
minum in dialysis fluids and the concomitant intake of was contaminated sulfuric acid, which was used to treat
aluminum-containing drugs. The highest occupational cane sugar. It was observed that some persons seemed
exposures occur among aluminum welders and pow- to have a “peculiar idiosyncracy” and “that many per-
der production workers. sons became ill who drank less beer than others who
Australian gold miners who inhaled aluminum were not affected” (Klatsky, 2006). The authors drew
powder in order to prevent silicosis had a slightly the conclusion that arsenic predisposed susceptible
increased cardiovascular mortality compared to other persons to develop alcoholic cardiomyopathy. Arsenic
gold miners [hazard ratio (HR) 1.02 per year of expo- intoxication has been associated with atypical ven-
sure, 95% confidence interval (CI) 1.00-1.04] (Peters tricular tachycardia (Goldsmith and From, 1980). Mice
et al., 2013). treated orally with NaAsO2 for 2 days developed lipid
Several studies of primary aluminum smelter work- peroxidation and abnormal ultrastructural changes in
ers reported an increased risk of ischemic heart and the cardiac tissue (Manna et al., 2008). These authors
cerebrovascular diseases (Rønneberg, 1995; Thériault summarized that arsenic exposure is associated with
et al., 1988). Increased risks were also found among direct myocardial injury, cardiac arrhythmias, and car-
potroom workers at the old Söderberg pots, as well as diomyopathy (Manna et al., 2008).
among workers using prebaked electrodes (Thériault A review of 11 cross-sectional studies identified an
et al., 1988). Slightly higher levels of fibrinogen were association between arsenic exposure and the preva-
observed among smelter workers at Söderberg pots lence of hypertension. The association was evident in
exposed to high levels of air pollutants compared to studies conducted both in areas with arsenic drink-
those working with prebaked electrodes and exposed ing water concentrations above 50 μg/L and in those
to lower levels of air pollutants (Sjögren et al., 2002). with drinking water concentrations below 50 μg/L.
16  Cardiovascular Disease 315

The evidence is suggestive of an association but is relationship for increasing categories of arsenic expo-
insufficient to infer a causal relationship because the sure (Axelson et al., 1978). In a cohort study of 3916
number of studies was small and no prospective stud- male smelter workers from the same smelter, slightly
ies were done (Abhyankar et al., 2012). increased mortality from IHD was found (SMR 1.07,
A Spanish ecological investigation in a population 95% CI 0.97-1.17) but there was no dose-response rela-
of 24.8 million people related arsenic concentrations tionship (Järup et al., 1989).
in drinking water during 1998-2002 to mortality for In summary, the consumption of beer contaminated
the period 1999-2003 to allow for a 1-year delay with with arsenic was associated with cardiomyopathy. A
respect to arsenic determinations. An arsenic concen- high concentration of arsenic in drinking water is asso-
tration above 10 μg/L was associated with an increased ciated with cardiovascular, including peripheral vas-
risk of CVD and CHD mortality in men as well as cular, disease. Some studies of occupationally exposed
women (Medrano et al., 2010). In the United States, primary smelter workers suggest a relationship
millions of persons are exposed to arsenic concentra- between arsenic exposure and CVD, although con-
tions above 10 μg/L and populations in many coun- founding exposures may contribute to the observed
tries (e.g. Bangladesh, China, India, Cambodia, Ghana, excess risk.
Argentina, and Mexico) are exposed to arsenic levels in
drinking water above 10 μg/L (Abhyankar et al., 2012).
A pooled analysis included groups exposed to 4 BERYLLIUM
arsenic concentrations above 50 μg/L in drinking
water. The pooled relative risks (RRs) for CVD, CHD, The worldwide use of beryllium is increasing and
stroke, and peripheral arterial disease were 1.32 (95% occupational beryllium exposure occurs in the aero-
CI 1.05-1.67), 1.89 (95% CI 1.33-2.69), 1.08 (95% CI space, nuclear, automotive, electronics, and telecommu-
0.98-1.19), and 2.17 (95% CI 1.47-3.20), respectively nication industries. Prolonged inhalation of beryllium
(Moon et al., 2012). may stimulate the acquired immune response to
Low levels of arsenic are common in copper ore all release mediators of chronic inflammation in the lung,
over the world, and primary copper smelter work- involving cellular and molecular components of innate
ers are exposed to arsenic, as well as to several other immunity, and it is this vicious cycle driven by beryl-
compounds such as silica dust, and sulfur dioxide. lium that results in the progressive impairment of lung
A cohort of 8047 white male workers from copper function, granuloma formation, and progression to
smelters in Montana exposed to arsenic trioxide was lung fibrosis (Sawyer and Maier, 2011). Absorption of
followed until 1963; increased mortality from heart beryllium from ingestion is very low (see Chapter 30
disease was observed [standardized mortality ratio “Beryllium”).
(SMR) 1.18, P < 0.01]. However, these smelter work- Several U.S. cohort studies of beryllium-exposed
ers were also exposed to sulfur dioxide and silica, and workers have reported increased mortality from heart
some also to lead (Lee and Fraumeni, 1969). A subco- diseases. A cohort of 3055 white male beryllium-
hort of 1800 men from the same smelter was followed exposed workers employed between 1942 and 1968
until 1978. In the group heavily exposed to arsenic was followed until 1975. An increased risk of heart dis-
(> 0.5 mg/m3), increased IHD mortality was observed ease was observed among workers employed before
(SMR 1.77, P < 0.01). A tentative dose-response was 1950 (SMR 1.16, P < 0.05). The risk of death from heart
observed when exposure to the highest level of arsenic disease was higher for workers exposed for more than
for at least 30 days was related to IHD mortality (Welch 5 years (SMR 1.29) than among workers exposed for
et al., 1982). The total cohort of 8045 arsenic-exposed less than 5 years (SMR 1.10) (Wagoner et al., 1980). The
copper smelter workers in Montana followed until estimated air concentrations of beryllium in the 1940s
1977 also had increased mortality from heart disease frequently exceeded 1000 μg/m3 in some of the indus-
(Lee-Feldstein, 1983). tries included (Ward et al., 1992).
A total of 2802 white males were included in a A U.S. cohort comprising 9225 males who had
cohort of copper smelter workers in Tacoma, Washing- worked between 1940 and 1969 was followed until
ton. After adjustment for the healthy worker survivor 1988. The cohort members had worked in plants
effect, significantly increased mortality from CVD was engaged in beryllium processing, including the extrac-
observed after high cumulative exposure to arsenic tion of beryllium hydroxide from beryl ore, the pro-
(Hertz-Picciotto et al., 2000). duction of beryllium oxide, pure beryllium metal, and
A case-referent study from a Swedish primary cop- beryllium copper alloy, and the machining of beryl-
per smelter showed significantly increased mortality lium-containing products. Mortality from IHD was
from CVD. There was also a significant dose-response significantly increased in the total cohort (SMR 1.08,
316 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

95% CI 1.01-1.14). Mortality from diseases of the arter- of cadmium were strongly positively associated with
ies, veins, and pulmonary circulation, which includes peripheral arterial disease. After multivariate adjust-
cor pulmonale, was increased in one plant (Lorain) ment (age, sex, race, education level, and smoking
(SMR 1.78, P < 0.05). Cor pulmonale is a heart disease status), subjects with peripheral arterial disease had
characterized by enlargement and hypertrophy of the 36% higher levels of cadmium in urine than subjects
right ventricle secondary to increased resistance and without peripheral arterial disease (Navas-Acien et al.,
high blood pressure in the pulmonary circulation. 2005). The results support a possible role for cadmium
Beryllium pneumoconiosis causes such an increased in atherosclerosis.
resistance in the pulmonary circulation (Ward et al., A 15-year follow-up study of urinary cadmium and
1992). The cohort was updated with quantitative expo- mortality among inhabitants of a cadmium-polluted
sure estimates, expanded to 9199 workers, and fol- area (Kakehashi River Basin) in Japan showed that the
lowed until December 2005. Increased mortality from mortality risk of subjects with high urinary cadmium
cor pulmonale was reported (SMR 1.17, 95% CI 1.08- (≥ 10 μg/g creatinine) after adjustment for age was
1.26). No risk estimate for mortality from IHD was higher than that of subjects with moderate urinary
reported (Schubauer-Berigan et al., 2011). cadmium (< 10 μg/g creatinine) in both sexes. When
To summarize, an increased risk of IHD was the subjects were divided into five groups accord-
observed in one study and increased risks of cor pul- ing to the amount of urinary cadmium (< 3, 3-5, 5-10,
monale were reported in two studies. A suggestive 10-20, and ≥ 20 μg/g creatinine), the mortality risk was
dose-response relationship was found for heart disease significantly increased among women with urinary
including both IHD and cor pulmonale. The increased cadmium ≥ 3 μg/g creatinine. The results suggested
risk may have been caused by the very high exposure that mortality risk ratio is increased in proportion to
levels (around 1000 μg/m3) prevalent before 1950. increases in the urinary cadmium concentration after
adjustment for age. When special causes of death
among subjects with high and moderate urinary cad-
5 CADMIUM mium were investigated, the mortality risk ratio for
heart failure was significantly increased in both sexes
Cadmium is widely spread in the environment. compared with those with moderate urinary cadmium
Exposure to humans occurs by ingestion of food con- (Nakagawa et al., 2006).
taminated with cadmium, by smoking, and in certain Another follow-up study among the inhabitants of
industrial settings. Smokers have approximately twice the cadmium-polluted Kakehashi River Basin area in
the body burden of cadmium compared to nonsmok- Japan showed excess mortality due to heart failure and
ers. In nonsmokers, the primary source of exposure cerebral infarction in both sexes, as well as nephritis
is food (Gallagher & Meliker, 2010). Exposure to cad- and nephrosis in men, among subjects with renal tubu-
mium has been associated with diseases of the cardio- lar dysfunction (urinary β2-microglobulin > 1000 μg/g
vascular system, bone, kidneys, and lungs. Cadmium creatinine) induced by cadmium in the environment.
and cadmium compounds are classified as carcinogens A significant increase in mortality risk for cerebral
(see also Chapter 18 “Carcinogenicity of Metal Com- infarction in men was observed during the first five-
pounds” and Chapter 32 “Cadmium” of this book). year observation period. The mortality risk for heart
The effects of cadmium on the cardiovascular system failure and cerebral infarction increased in proportion
have been suggested to be a result of its action on to increased urinary β2-microglobulin in both sexes
the cardiovascular endothelium and smooth muscle, (Nishijo et al., 2006).
mediated by the generation of a proinflammatory In a Belgian study, subjects were randomly
state, endothelium dysfunction, increased oxidative recruited from an area contaminated with cadmium
stress, and downregulation of nitric oxide production and a reference area with lower exposure to cadmium,
(Agarwal et al., 2011). This chapter describes investiga- and blood cadmium, urinary cadmium, and mortal-
tions in the general population and also occupational ity were followed. At baseline, blood cadmium (14.6
mortality studies. versus 10.2 nmol/L) and urinary cadmium (14.1 ver-
Blood levels of cadmium were associated with an sus 8.6 nmol/24 hr) were higher in subjects who died
increased prevalence of peripheral arterial disease than in survivors. However, there was no relationship
(defined as a blood pressure ankle-brachial index < 0.9 between cadmium levels and cardiovascular mortality
in at least one leg) in a representative sample (cross- (Nawrot et al., 2008).
sectional analysis) of the general U.S. population In a cross-sectional study of U.S. adults, the cad-
(Navas-Acien et al., 2004). The results were similar to mium level in blood, but not in urine, was associ-
a study in the same population in which urinary levels ated with a modest elevation in blood pressure. The
16  Cardiovascular Disease 317

average difference in systolic and diastolic blood pres- was associated with increased cardiovascular mortal-
sure between participants in the 90th versus the 10th ity and increased incidence of CVD. After adjusting for
percentile of the blood cadmium distribution were sociodemographic and cardiovascular risk factors, the
1.36 mm Hg (95% CI −0.28 to 3.00) and 1.68 mm Hg HRs (comparing the 80th to the 20th percentile of urine
(95% CI 0.57-2.78), respectively. There was no associa- cadmium concentrations) were 1.43 for cardiovascular
tion between cadmium levels and the prevalence of mortality (95% CI 1.21-1.70) and 1.34 for CHD mor-
hypertension (Tellez-Plaza et al., 2008). tality (95% CI 1.10-1.63). The corresponding HRs for
Cadmium in the blood and urine in the general U.S. incident CVD, CHD, stroke, and heart failure were 1.24
population was positively associated with the risk of (95% CI 1.11-1.38), 1.22 (95% CI 1.08-1.38), 1.75 (95%
having a history of stroke or heart failure. After adjust- CI 1.17-2.59), and 1.39 (95% CI 1.01-1.94), respectively
ing for demographic and cardiovascular risk factors, (Tellez-Plaza et al., 2013a).
a 50% increase in blood cadmium corresponded to a In a systematic review by Tellez-Plaza et al. (2013b),
35% increased odds of a history of stroke (OR 1.35, 95% the authors state that, together with experimental evi-
CI 1.12-1.65) and a 48% increased odds for a history of dence, their review (12 studies were included) sup-
heart failure (OR 1.48, 95% CI 1.17-1.87). A 50% increase ports an association between cadmium exposure and
in urinary cadmium corresponded to a 9% increase in CVD, especially for CHD. The number of studies with
odds for a history of stroke (OR 1.09, 95% CI 1.00-1.19) stroke, heart failure, and peripheral artery disease end-
and a 12% increase in odds for a history of hyperten- points was small. Overall, the pooled RRs for CVD,
sion (OR 1.12, 95% CI 1.03-1.20) (Peters et al., 2010). CHD, stroke, and peripheral artery disease were 1.36
In a review by Alissa and Ferns (2011), the authors (95% CI 1.11-1.66), 1.30 (95% CI 1.12-1.52), 1.18 (95% CI
discuss a possible association between cadmium expo- 0.86-1.59), and 1.49 (95% CI 1.15-1.92), respectively. The
sure and hypertension, diabetes, myocardial infarc- pooled RRs for CVD in men, women, and never smok-
tion, early atherosclerotic vessel wall thickening, and ers were 1.29 (95% CI 1.12-1.48), 1.20 (95% CI 0.92-1.56)
CVD, although there may be confounding exposures, and 1.27 (95% CI 0.97-1.67), respectively.
for example coexposure to other metals and residual In a cohort mortality study in Great Britain almost
confounding from smoking habits. 7000 male cadmium-exposed workers from five indus-
Cadmium levels in blood and urine were associated tries (primary production, copper-cadmium alloys,
with the prevalence and future growth of atheroscle- silver-cadmium alloys, pigments and oxide, and stabi-
rotic plaques in the carotid arteries (Fagerberg et al., lizers), who were exposed for more than 1 year between
2012), and with low ankle-brachial index as a measure 1942 and 1970, were followed with regard to mortality
of peripheral artery disease (Fagerberg et al., 2013), in a to the end of 1984. Compared with the general popu-
cohort of 64-year-old Swedish women, after adjusting lation, there was no significant excess of death due to
for smoking and other cardiovascular risk factors. hypertensive disease (SMR 1.19, 95% CI 0.85-1.52) and
A prospective study in the general U.S. population the SMR for deaths from cerebrovascular disease was
(8989 participants who were ≥ 20 years of age) showed significantly lower than expected (SMR 0.77, 95% CI
that cadmium levels in the blood and urine were asso- 0.66-0.89) (Kazantzis et al., 1988).
ciated with future CVD mortality, heart disease mor- In a mortality study of 347 male copper-cadmium
tality, and for cadmium levels in urine also with CHD alloy workers in Great Britain (all subjects first
mortality, after adjusting for sociodemographic and employed in the period 1922-1978 and for a minimum
CVD risk factors, including smoking status, recent period of 1 year) were investigated for the period 1946-
smoking dose, and cumulative smoking dose. The 1992. Compared with the general population, there
adjusted HRs for blood and urine cadmium, compar- was no excess of mortality from diseases of the circu-
ing the 80th to the 20th percentiles of cadmium distri- latory system (SMR 1.03, 95% CI 0.83-1.27) (Sorahan
butions, were 1.69 (95% CI 1.03-2.77) and 1.74 (95% CI et al., 1995).
1.07-2.83) respectively, for CVD mortality; 1.98 (95% CI In a mortality study of 869 Swedish battery work-
1.11- 3.54) and 2.53 (95% CI 1.54-4.16) for heart disease ers exposed to nickel hydroxide and cadmium oxide
mortality; and 1.73 (95% CI 0.88-3.40) and 2.09 (95% (employed for at least 1 year between 1940 and 1980),
CI 1.06-4.13) for CHD mortality. The population attrib- the workers were followed up until 1992. Compared
utable risks associated with the 80th percentile of the with the general population, the SMRs for IHD were
blood (0.80 μg/L) and urine (0.57 μg/g creatinine) cad- 1.16 (95% CI 0.96-1.40) in males and 0.75 (95% CI 0.25-
mium distributions were 7.5% and 9.2%, respectively, 1.76) in females, and the SMRs for cerebrovascular
for CVD mortality (Tellez-Plaza et al., 2012). disease were 0.78 (95% CI 0.47-1.21) in males and 1.34
A prospective cohort study of 3348 American Indian (95% CI 0.37-3.43) in females, after adjusting for smok-
adults 45-74 years of age showed that urine cadmium ing habits (Järup et al., 1998).
318 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

A cohort of 1462 male workers at a UK tin smelter mortality due to respiratory cancer indicates decreased
(who had been employed for at least 1 year between exposure to a very potent carcinogen. The author sus-
1967 and 1995) were followed up until 2001. Mortality pects calcium chromate to be the carcinogenic agent in
from IHD showed a deficit compared with the national these old chromate-producing plants. However, it was
population (SMR 0.96, 95% CI 0.82-1.11) and with unknown whether all of the excess deaths were related
the regional population (SMR 0.94, 95% CI 0.80-1.09) to exposures to chromate production (Enterline, 1974).
(Binks et al., 2005). A total of 1737 male workers were studied in a
A cohort of 2422 male workers (employed for three New Jersey chromium pigment factory. The pigments
or more years between 1946 and 1996) exposed to arse- contained both lead and zinc chromates. IHD mor-
nic, cadmium, and other substances at a copper mine tality was 0.85 (95% CI 0.71-1.00) (Hayes et al., 1989).
and smelter in Copperhill, Tennessee, were followed Another cohort originating from New Jersey com-
up until 2000. Based on national and local county prised 3408 workers who had worked between 1937
comparisons, there was no evidence of an increased and 1971 in former chromate-producing plants in the
mortality risk from cerebrovascular disease, all heart northern part of the state. The proportionate mortality
disease, or hypertension (Marsh et al., 2009). ratios regarding arteriosclerotic heart disease was 0.97
In summary, the data support an association of for white men, 0.90 for black men, and 1.19 (95% CI
cadmium exposure with the occurrence of CVD and 0.73-1.83) for white women. However, a weakness of
CHD. There is also a possible association with periph- the study is the inability to distinguish between pro-
eral artery disease, stroke, and heart failure. Several duction workers and management staff (Rosenman
occupational mortality studies showed no association and Stanbury, 1996).
between cadmium exposure and CVD compared with A cohort of 482 former chromate production work-
the general population. ers from Painesville, Ohio, USA, was followed until
1997. Forty-one percent started working at the chro-
mate plant during the 1940s, and 59% started before
6 CHROMIUM 1955. The average airborne concentration of hexava-
lent chromium in the indoor operating areas of the
The diet is the main source of chromium among plant from 1943 to 1948 was 720 μg/m3, from 1957
humans who are not occupationally exposed. Occupa- to 1964 was 270 μg/m3, and from 1965 through 1972
tional chromium exposure occurs in the manufactur- was 39 μg/m3. SMR for diseases of the heart was 1.13
ing of chromates and ferrochromium, in chromium (95% CI 0.93-1.36) based on Ohio reference rates and
plating, and in stainless steel welding. 1.21 (95% CI 0.99-1.46) based on U.S. reference rates.
In the early 1960s, the University of Pittsburgh was The corresponding SMRs for other circulatory dis-
contracted to study mortality in males born in 1890 eases were 1.43 (95% CI 0.96-2.04) and 1.51 (95% CI
or later and employed in three chromate-producing 1.02-2.16), respectively (Crump et al., 2003; Luippold
plants between 1937 and 1940; they were followed et al., 2003).
for mortality until 1961. Arteriosclerotic heart dis- A chromate chemicals manufacturing facility in
ease mortality was 1.93 in 1941-45 and 0.64 in 1956-60 Castle Hayne, North Carolina, was built in 1971 and
(Table 1). During the initial period (1941-1945) death designed to reduce the high levels of chromium expo-
rates were very high for some causes. The decline in sure found at most older facilities. A cohort comprised
398 workers employed between 1971 and 1989. More
TABLE 1  Mortality among Chromate Workers, Working than 99% of the measurements of hexavalent chromium
between 1937 and 1940, Followed until 1960 based on personal sampling were below 50 μg/m3. Forty-
five employees had previously worked at the plant in
1941–1945 1946–1950 1951–1955 1956–1960
Painesville, Ohio. The SMR for all heart diseases was
Cause of
death Obs SMR Obs SMR Obs SMR Obs SMR 0.65 (90% CI 0.26-1.37) based on only five cases and the
mortality experience of the surrounding eight North
All 59 1.58 55 1.26 75 1.35 57 1.00 Carolina counties (Pastides et al., 1994).
Respiratory 16 29.1 19 15.7 19 7.92 15 4.75 A cohort study of employees was formed from two
cancer
different U.S. plants. Exposure levels to hexavalent
Arterioscle- 11 1.93 12 1.13 18 1.03 13 0.64
rotic heart
chromium at both plants were very low after major
disease process changes from high-lime to no-lime processes.
The geometric mean of personal air concentrations of
Obs, observed; SMR, standardized mortality ratio. Cr(VI) remained much less than 1.5 μg/m3 for most
Source: Enterline, 1974. years at both plants. The range of annual means was
16  Cardiovascular Disease 319

0.36 to 4.36 μg/m3 and the highest personal air sam- role with cobalt in the development of this disease
pling values were generally less than 10 μg/m3. This (Lauwerys and Lison, 1994; Seghizzi et al., 1994).
study examines the mortality experience of two post- Cardiomyopathy was also described in two workers
change cohorts of chromate production employees exposed to heavy dust concentrations from cobalt-
constituting the current U.S. chromium chemical containing ores for 2 months and 2 years, respectively
industry. The SMR for IHD was 0.55 (95% CI 0.18-1.28) (Jarvis et al., 1992). Cardiomyopathy was also reported
based on five cases (Luippold et al., 2005.) after exposure to hard-metal dust in a man responsible
A total of 2298 chromate-producing workers were for weighing and mixing tungsten carbide and cobalt
identified from three different factories (Bolton, Ruther- powder (Kennedy et al., 1981).
glen and Eaglescliffe) in the United Kingdom. They The bioavailability of cobalt increases in the pres-
had worked for at least 1 year between 1950 and 1976 ence of tungsten carbide. Endotracheal instillation of
and were followed until 1988. During this follow-up tungsten carbide-cobalt mixture in rats causes severe
no significant increases were observed in the different alveolitis and fatal pulmonary edema, whereas the
factories regarding IHD (SMR 1.03-1.12) or cerebrovas- corresponding dose of cobalt powder alone causes
cular disease (SMR 1.06-1.30) (Davies et al., 1991). a moderate inflammatory response (Lauwerys and
Two cohorts from two German chromate-producing Lison, 1994).
factories comprised 1417 workers; they were fol- Mice exposed to 30 mg/m3 cobalt sulfate heptahy-
lowed until 1988. Both factories were originally high- drate for 90 days developed arteritis in the main cor-
lime facilities. The plant in Leverkusen completed onary artery (2/17 compared to controls 0/18). In a
the changeover to a no-lime process in 1958 and the long-term inhalation study of exposure to 3.0 mg/m3
plant in Uerdingen completed the changeover in 1964. cobalt for 24 months, 2/48 animals developed arte-
Mortality due to CVD was 1.16 (95% CI 0.97-1.39) in ritis in coronary arteries versus 0/46 in controls. The
Leverkusen and 0.96 (95% CI 0.78-1.15) in Uerdin-
­ number of animals with arteritis in the renal arteries
gen (Korallus et al., 1993). In a later study, 901 work- was 5/48 versus 1/46. Thus, there was a significantly
ers exposed only after the changeover formed a new increased risk when all groups were taken into account
cohort, which was followed until 1998. Exposure (Moyer et al., 2002).
declined from the mid-1970s based on chromium lev- In a cross-sectional study of 30 cemented tungsten
els in urine samples. The SMR for diseases of the heart carbide workers with a mean exposure duration of 9.9
was 0.66 (95% CI 0.45-0.93) and for other diseases of years, a weak but significant inverse correlation was
the circulatory system was 1.00 (95% CI 0.57-1.63) (Birk found between duration of exposure and resting left
et al., 2006), thus suggesting a reduced risk. ventricular function. Nine workers with abnormal
A Japanese cohort study of 896 male workers chest X-ray findings had relatively lower exercise right
manufacturing chromium compounds observed an ventricular ejection fractions. Diminished right ven-
increased SMR for lung cancer (9.5) but no increases tricular reserve was probably due to fibrotic lung dis-
for heart disease (SMR 0.6) and cerebrovascular dis- ease and early cor pulmonale. The authors concluded
ease (SMR 1.0) (Satoh et al., 1981). that although overt left ventricular dysfunction was
In summary, one study of chromate-producing not present, prolonged exposure to industrial cobalt
workers from the USA has shown decreasing trends may have a weak cardiomyopathic effect (Horowitz
of cardiovascular mortality that may be associated et al., 1988).
with improvements in the working environment and No increased mortality risk of all circulatory dis-
decreasing exposures. These observations may indi- ease or IHD were observed in the largest cohort of
cate a relationship between hexavalent chromium French hard-metal workers comprising totally 7459
exposure and CVD. Hexavalent chromium exposures men and women. The SMRs were 0.88 (95% CI 0.75-
below 50 μg/m3 were not associated with heart disease. 1.03) and 0.93 (95% CI 0.71-1.19), respectively (Moulin
et al., 1998).
A cohort of 3163 male Swedish hard-metal workers
7 COBALT was followed from 1951 to 1982. In the total cohort,
the SMR for IHD was 0.99 (95% CI 0.80-1.21). In a sub-
In the early 1960s cobalt salts, chloride or sulfate, group with estimated high cobalt exposure and long-
were added to some brands of beer in order to stabi- term (> 10 years) exposed workers, a higher SMR was
lize the foam; as a result of this intervention, heavy observed (SMR 1.69, 95% CI 0.96-2.75) (Hogstedt and
beer drinkers developed cardiomyopathy, which led Alexandersson, 1990).
to a high mortality rate (Morin et al., 1971). It is likely Thus, cardiomyopathy has been induced by the
that poor nutrition and ethanol played a synergistic intake of cobalt-containing beer and after inhalation
320 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

of high concentrations of dust from cobalt-containing Dietary intake of iron was assessed at baseline by
ores. Signs of cor pulmonale have been observed as a a questionnaire in a cohort study comprising 58,615
result of fibrotic lung disease induced by inhalation of healthy Japanese. Dietary intake of total iron was posi-
hard-metal dust. The majority of cohorts comprising tively associated with mortality from total and isch-
cobalt-exposed hard-metal workers do not report an emic stroke and total CVD in men but not in women
increased risk of IHD. However, one study suggests an (Zhang et al., 2012a).
excess in long-term exposed workers. Several occupations are associated with exposure
of airborne iron-containing particles e.g. welders and
iron foundry workers. However, these workers are
8 IRON exposed to other metals and gaseous air pollutants in
addition to iron, which may confound possible asso-
Iron is an essential metal for the human body. ciations between iron exposure and outcomes.
Almost all cells employ iron as a cofactor for funda-
mental biochemical activities, such as oxygen trans-
8.1 Welders
port, energy metabolism, and DNA synthesis. The vast
majority of body iron is distributed in the hemoglo- Welding processes generate both respirable particles
bin of red blood cells and developing erythroid cells. and gases. Iron is the major particulate component in
Iron is absorbed from the diet mainly in two forms of most steel-welding operations (Flynn and Susi, 2010).
iron; heme and nonheme (inorganic) iron. Humans are An increase in the inflammatory marker CRP was
unable to excrete iron actively, and the concentration observed among welders from an apprentice weld-
in the body is regulated at the site of iron absorption in ing school who mainly worked in low-alloy steel for 1
the proximal small intestine (Wang and Pantopoulos, day, with a mean exposure of 1.7 mg/m3 measured as
2011; Zimmermann and Hurrell, 2007). Welders inhale particulate matter with an aerodynamic mass median
iron oxide and the lungs of welders exposed continu- diameter of 2.5 μm (PM2.5) (Kim et al., 2005).
ously for 18 years may contain between 30 and 2000 mg Increased mortality from IHD was observed
(Kalliomäki et al., 1978). among welders, despite the fact that the death rates
Iron overload is the accumulation of excess iron of these welders were compared with those of the
in different organs as a result of increased intestinal general population (which also includes sick and dis-
absorption, parenteral administration, or increased abled people unable to work); consequently, the risk
dietary intake. During iron overload, transferrin, the estimates may underestimate the true risk (Moulin
carrier of iron in the circulation, which is normally et al., 1993; Newhouse et al., 1985). Some cohort stud-
around 30% saturated, becomes fully saturated, and ies of welders did not observe any significant differ-
toxic non-transferrin-bound iron species appear in the ences in diseases of the circulatory system (Becker,
blood. The heart is one of the main organs affected by 1999; Simonato et al., 1991); in one study a signifi-
excess iron accumulation. The term iron overload car- cantly decreased risk was observed (Beaumont and
diomyopathy refers to cardiomyopathy resulting from Weiss, 1980). Later studies were better designed and
iron accumulation in the myocardium. This condition included both fatal and nonfatal cases of disease. In
may be caused by genetically determined disorders a Danish study, 8376 male metal workers answered
of iron metabolism or by multiple blood transfu- a questionnaire about their welding experience and
sions. This myopathy comprises different forms of lifestyle. A cohort of 5866 welders was followed from
cardiac dysfunction secondary to the iron deposition. the start of 1987 until the end of 2006. Information
Iron overload cardiomyopathy accounts for one-third of CVD was retrieved from the Danish National
of the deaths in hereditary hemochromatosis and is Patient Registry. Increased risks were observed for
the leading cause of mortality in thalassemia major acute myocardial infarction [standardized incidence
(­Kremastinos and Farmakis, 2011). ratio (SIR) 1.1, 95% CI 1.0-1.2], chronic IHD (SIR 1.2,
The total iron-binding capacity (TIBC) represents 95% CI 1.1-1.3), and cerebral infarction (SIR 1.2, 95%
the level of transferrin, which is usually inversely CI 1.1-1.4) (Ibfelt et al., 2010). A Swedish record-
correlated to iron stores. In a meta-analysis compris- linkage study found an increased risk of acute myo-
ing seven studies, high TIBC levels were associated cardial infarction both among male (HR 1.2, 95% CI
with a decreased risk of coronary artery disease. The 1.1-1.3) and female (HR 1.3, 95% CI 1.1-1.6) welders.
association persisted when only prospective studies An increased risk was also observed for other IHDs
were included in the analysis (Kang et al., 2012). Thus, among males (HR 1.2, 95% CI 1.1-1.2) but the risk was
increased iron stores seem to be associated with an not as pronounced among females (HR 1.1, 95% CI
increased risk of CHD. 0.98-1.3) (Wiebert et al., 2012).
16  Cardiovascular Disease 321

In a cross-sectional study, 97 welders were com- disease mortality was related to increasing duration of
pared with 91 nonwelding controls (mean ages 35.7 exposure (Michaels et al., 1991; Steenland et al., 1992).
and 35.4 years, respectively). Male welders had signifi- Navas-Acien and coworkers (2007) concluded that the
cantly higher serum concentrations of iron than con- evidence was sufficient to infer a causal relationship
trols, but this association was not found among female between lead exposure and hypertension. They also
welders (Lu et al., 2005). concluded that the evidence was suggestive of, but not
In summary, iron overload can cause cardiomyopa- sufficient to infer, a causal relationship between lead
thy. Several studies have also observed an association exposure and clinical cardiovascular outcomes.
between iron stores and an increased risk of CHD. After the review by Navas-Acien and coworkers
Welders are generally exposed to small particles of iron (2007), some further studies have been published. A
but also to many other particles of metal or nonmetal Russian study in printing workers comprised 1423 men
origin. Several studies of welders support the associa- and 3102 women. An increased risk of IHD was found
tion between airborne particle exposure and the occur- for male typecasting machine operators (SMR 1.29, 95%
rence of CVD, and one study observed an increase in CI 1.08-1.56). Information on exposure was based on
an inflammatory marker. an industrial hygiene survey and exposure duration
in years. However, no dose-response relationship was
observed. In male or female manual typesetters and
9 LEAD linotype operators no increased risks of cerebrovascu-
lar disease were observed (Ilychova and Zaridze, 2012).
The general population may be exposed to lead from Among 868 totally environmentally exposed men,
leaded gasoline, lead in paint, and industrial emis- lead levels in bone (patella and tibia) were measured
sions. Occupational exposures originate mainly from and the average follow up was 8.9 years. The highest
smelters, refineries, and battery and paint production. tertile of lead bone levels was compared with the low-
Several mechanisms have been reported to explain the est. This comparison resulted in significantly increased
observed increases of blood pressure associated with mortalities for both CVD (HR 2.5, 95% CI 1.1-5.6) and
lead exposure, including impaired renal function, the IHD (HR 8.4, 95% CI 1.3-54.4) (Weisskopf et al., 2009).
induction of oxidative stress and inflammation, and In 2012 the U.S. National Toxicology Program (NTP)
endothelial dysfunction (Navas-Acien et al., 2007). reviewed the health effects of low-level lead expo-
A meta-analysis focusing on blood lead and blood sure. The NTP evaluation declared that there is suffi-
pressure comprised a total of 31 studies. An association cient evidence that blood lead levels below 10 μg/dL
between blood lead and blood pressure was found in (0.5 μmol/L) are associated with small but detectable
both men and women. A twofold increase in blood lead increases in blood pressure and risk of hypertension.
concentration was associated with a 1.0-mm Hg rise in The NTP concluded that there is limited evidence of
systolic blood pressure (95% CI 0.5-1.4 mm Hg) and a CVD mortality at blood levels below (0.5 μmol/L).
0.6-mm Hg rise in diastolic blood pressure (95% CI 0.4- Lead levels in bone reflect long-term lead exposure,
0.8 mm Hg) (Nawrot et al., 2002). An increase in tibia lead and these levels were more consistently associated
by 10 μg/g bone was associated with a 0.26 mm Hg rise in with hypertension, CVD and mortality (NTP, 2012).
systolic blood pressure (95% CI 0.02-0.5 mm Hg) and the In summary, there is strong evidence for an asso-
summary odds ratio per 10 μg/g bone for hypertension ciation between lead exposure and both hypertension
was 1.04 (95% CI 1.01-1.07) (Navas-Acien et al., 2008a). and CVD. Some studies in occupationally exposed
Navas-Acien and coworkers (2007) reviewed the workers showed a relationship between lead exposure
literature on the relation between lead exposure and and cerebrovascular diseases after long-term exposure.
CVD. The review included 12 studies of the general It has been estimated that 4% of the total global bur-
population: five prospective cohort studies, one cross- den of IHD is due to lead exposure (annually, 674,000
sectional study, and six case-control studies. Lead deaths and 13.9 million disability-adjusted life-years,
exposure was positively associated with clinical car- which is the sum of years of life lost due to death and
diovascular endpoints in all studies. The evaluation years of life with disability) (Lim et al., 2012).
included 18 studies with occupational exposures from
different industrial settings: battery, ceramic, refin-
ery, and smelter industries. Most were cohort studies 10 MAGNESIUM
and used external comparisons to the general popu-
lation to derive SMRs. Relative risk estimates varied Magnesium is the fourth most common cation in the
widely. In two of the three studies that reported asso- body and the second most abundant intracellular cat-
ciations by duration of employment, cerebrovascular ion after potassium. Magnesium plays an essential role
322 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

as an enzymatic cofactor in a number of biochemical stronger for women than for men. Further adjustment
pathways, including energy production from adenos- for diabetes status and systolic blood pressure elimi-
ine triphosphate (ATP). This cation has a direct effect nated the relation for men. However, the authors dis-
on the Na+, K+-ATPase pump in both cardiac and cuss whether this was an appropriate way to adjust
nerve tissues. Magnesium is a cofactor in more than for two confounders or whether an overadjustment
300 enzyme systems in human cells and has a piv- had been made for factors within a causal pathway if
otal position in normal myocardial physiology. Pos- low magnesium concentrations had contributed to the
sible sites of action include vascular smooth muscle, development of diabetes and hypertension (Liao et al.,
thrombocytes, and myocardial cells (Elin, 1987; Marx 1998). The cohort was followed further until 2004.
and Neutra, 1997). Oral magnesium acts as a natural Serum magnesium was inversely associated with isch-
channel blocker, increases nitric oxide, improves endo- emic stroke incidence (Ohira et al., 2009). In a separate
thelial dysfunction, and induces direct and indirect analysis, 7731 participants free of hypertension were
vasodilation (Houston, 2011). Investigations of animal followed for 6 years. The results from this study sug-
and cellular responses to magnesium deficiency have gest that low magnesium may play a modest role in
found evidence of complex proinflammatory path- the development of hypertension (Peacock et al., 1999).
ways that may lead to better understanding of inflam- In 2002, a meta-analysis of 20 studies detected
matory processes (Weglicki, 2012). dose-dependent blood pressure reductions following
The largest proportion of magnesium intake origi- magnesium supplementation. However, the authors
nates from food; the estimated intake from drinking recommended further adequately powered trials with
water is less than 5-10% (Rosenlund et al., 2005). A sufficiently high doses of magnesium supplementa-
low correlation was observed between dietary mag- tions to confirm this relationship (Jee et al., 2002).
nesium intake and serum levels of magnesium (Ohira Within the Cochrane Collaboration, magnesium
et al., 2009). The serum magnesium concentration cor- supplementation was reviewed for the management
relates with the intracellular magnesium level in red of primary hypertension in adults. A total of 12 trials
blood cells (Ryzen et al., 1989), but serum magnesium enrolling 545 persons compared magnesium with pla-
accounts for less than 1% of the whole body burden of cebo and measured blood pressure 8 weeks to 6 months
magnesium (Elin, 1987). later. On average, people receiving extra magnesium
Rodríguez-Móran and coworkers (2011) reviewed achieved slightly lower diastolic blood pressure at the
epidemiological studies and clinical trials regarding end of the trials. However, most trials included were
type 2 diabetes mellitus (T2DM). The body of evidence of poor quality, so the results may not be reliable. The
from epidemiological studies showed a strong inverse review did not find robust evidence that oral magne-
relationship between dietary magnesium intake and sium supplementation reduces high blood pressure in
the occurrence of T2DM. Magnesium is an important adults (Dickinson et al., 2006, 2009).
component of unprocessed foods such as whole grains, The Cochrane Heart Group (Li et al., 2007, 2009)
nuts, and green leafy vegetables. Randomized clinical evaluated the effect of intravenous magnesium on lim-
trials evaluating the benefits of magnesium supple- iting damage to the heart muscle, preventing serious
mentation on glucose levels in individuals with T2DM arrhythmias, and reducing the risk of death in heart
are scarce and controversial. attack patients. A large well-designed trial with 58,050
The intake of dietary magnesium was assessed participants did not demonstrate any beneficial effect
in 58,615 Japanese men and women. Dietary magne- of intravenous magnesium, contradicting earlier meta-
sium intake was inversely associated with mortality analyses of smaller trials (ISIS-4, 1995). This review
from hemorrhagic stroke in men and with mortality included 26 clinical trials that had randomly assigned
from total and ischemic strokes, CHD, heart failure, heart attack patients to receive either intravenous mag-
and total CVD in women (Zhang et al., 2012b). Previ- nesium or an inactive substance (placebo). This review
ous studies of magnesium dietary intake and ischemic did not support the continued use of intravenous mag-
stroke have observed the same inverse relation among nesium (Li et al., 2007, 2009).
women in the Nurses’ Health Study (Iso et al., 1999) Magnesium sulfate is a neuroprotective agent that
and among men from the USA (Ascherio et al., 1998). may hypothetically improve the outcome of aneurys-
The Atherosclerosis Risk in Communities (ARIC) mal subarachnoid hemorrhage by reducing the occur-
study comprised around 13,000 participants between rence or improving the outcome of delayed cerebral
1987 and 1989 in four U.S. communities. The partici- ischemia. In a clinical trial, 606 patients were assigned
pants were reexamined between 1990 and 1995. Low to the magnesium group and 597 patients were
serum magnesium values were associated with an assigned to the placebo group. Intravenous magne-
increased incidence of CHD. This association was sium sulfate did not improve the clinical outcome after
16  Cardiovascular Disease 323

aneurysmal subarachnoid hemorrhage. In a meta- with hypertension in this representative sample of the
analysis of six previous studies the same pattern was Korean adult population (Lee and Kim, 2011). A simi-
observed (Dorhout Mees et al., 2012). lar result was found in males from India (Taneja and
Inhalation exposure of magnesium has been studied Mandal, 2007). A U.S. cross-sectional study comprised
much less than magnesium exposure through drink- 639 older men in the Normative Aging Study (NAS).
ing water and dietary intake. The use of hydrated Toenail concentrations of several metals including
magnesium carbonate hydroxide (magnesia alba) for manganese were assessed. Manganese was negatively
drying the hands is an important source of airborne but not significantly associated with blood pressure.
particulate matter in indoor climbing halls. Mean val- After adjusting for other metals, an increase in man-
ues for PM10 in these halls were generally in the order ganese was more strongly associated with decreased
of 0.2-0.5 mg/m3. Periods of high activity were associ- blood pressure (Mordukhovich et al., 2012).
ated with levels of between 1 and 4 mg/m3 (Weinbruch Manganese-exposed ferroalloy workers had
et al., 2008). decreased systolic blood pressure (Saric and Hrustic,
Metal fume fever is an acute disability that occurs 1975); in another investigation, ferroalloy manufactur-
after inhalation of fumes from a metal heated to a ers had higher systolic and diastolic blood pressure
temperature above its melting point. Zinc, copper, than nonexposed referents (Cowan et al., 2009).
and magnesium are the most common of these metals Manganese superoxide dismutase (MnSOD;
(Hunter, 1978). Adding magnesium to molten iron has encoded by SOD2) is a member of the superoxide
induced fever in foundry workers (Hartmann et al., dismutase family, the members of which have anti-
1983). Investigations have not identified a relationship oxidant capacity. MnSOD may be the most important
between occupational magnesium exposure and CVD. member of the SOD family, as it resides in mitochon-
In summary, an inverse relationship has been dria, where cellular respiration takes place. One study
observed between dietary magnesium intake and the demonstrated that a SOD2 polymorphism (Ala16Val)
occurrence of diabetes and stroke. There is a low cor- may be associated with the development of T2DM in
relation between dietary magnesium intake and serum Japanese-Americans (Nakanishi et al., 2008). However,
levels of magnesium. Low serum magnesium may be manganese per se is not necessarily involved in this
associated with an increased risk of hypertension and observed effect.
CHD. Clinical studies into intravenous magnesium Rodier (1955) made an interesting observation when
treatment after a heart attack or aneurysmal subarach- studying manganese-exposed Moroccan miners. The
noid hemorrhage did not support beneficial effects of ore was very rich in manganese, neurological symp-
this regimen. toms were common among the underground miners,
and 150 cases of poisoning were reported. Each year
approximately 3.7% of the workers were affected by
11 MANGANESE pneumonia. Death from heart failure was observed
to occur after a week with pneumonia or occurred
Manganese is absorbed by ingestion and inhalation suddenly in patients regarded as cured. Manganese
after occupational exposure. Manganese ions may also pneumonia was associated with a marked rise in the
be injected intravenously in order to use their strongly erythrocyte sedimentation rate among the miners.
paramagnetic properties in magnetic resonance imag- The sedimentation rate can be influenced by several
ing. Cardiovascular magnetic resonance is an impor- serum proteins, for example, fibrinogen, haptoglobin,
tant diagnostic tool in cardiology. Manganese ions immunoglobins, and ceruloplasmin. Increased fibrino-
accumulate inside myocytes and enable better imag- gen concentration is an established risk factor for IHD
ing. Fifteen healthy volunteers were given manganese (Danesh et al., 2005) and has also been associated with
(MnCl2) intravenously. There was a slight increase in sudden death (Kucharska-Newton et al., 2009). In
systolic pressure and heart rate after 3 and 4 minutes another study of workers producing oxides and salts
of the infusion, with normalization of these parameters of manganese, the white blood cell count was higher
thereafter. On follow-up 6 months after investigation, among exposed workers than among referents when
no volunteer reported any neurological or cardio- smoking habits were taken into account (Roels et al.,
logical adverse events or symptoms (Fernandes et al., 1987). An increased white blood cell count is associated
2011). with an increased risk of CHD (Wheeler et al., 2004).
In a Korean National Health and Nutrition Exami- In a study of Norwegian ferromanganese-silico-
nation Survey (KNHANES), a total of 1991 partici- manganese furnace workers, three deaths due to
pants were included in cross-sectional design. Blood pneumonia occurred during active occupational expo-
manganese concentration was positively associated sure (Hobbesland et al., 1997a); increased mortality
324 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

from sudden death was also observed (Hobbesland other diseases of the heart, respectively (Boffetta et al.,
et al., 1997b). However, the furnace workers were also 2001). No conclusion can be drawn regarding mercury
exposed to other agents such as carbon monoxide exposure and circulatory diseases.
and heat. In 2002 the first results were presented from the
In summary, studies addressing the relation between Health Professionals Follow-Up Study (HPFS), a cohort
manganese exposure and blood pressure revealed con- study comprising 33,737 men who supplied toenail
flicting results and studies of other CVDs were very clippings. The cohort was followed from 1987 to 1992.
scarce. Dentists had higher levels of mercury in their toenails
(0.91 μg/g) compared with nondentists (0.45  μg/g).
The mercury levels were not significantly associated
12 MERCURY with the risk of CHD after adjustment for age, smok-
ing, and other risk factors (Yoshizawa et al., 2002). The
Vapor inhalation is the most important occupational U.S. HPFS cohort was followed further and a female
exposure route for mercury, typically occurring in mer- cohort from the Nurses’ Health Study was added. Mer-
cury mining and in chloralkali factories. In the general cury levels were derived from toenail clippings from
public, fish consumption is an important source of both studies. No increased risk of CHD, stroke, or
methylmercury exposure. total CVD was observed at higher mercury levels. The
Inhalation of large doses of elemental mercury median mercury concentration was 0.23 μg/g among
vapor results in an influenza-like illness (Gerstner and cases with CVD and 0.25 μg/g among control partici-
Huff, 1977). pants (Mozaffarian et al., 2011).
Increased cardiovascular mortality was found In population-based investigations, mercury expo-
among 1190 male workers exposed to inorganic mer- sure originates mainly from fish consumption. In
cury from eight Swedish chloralkali plants (when this a population-based nested case-control study from
cohort was compared with national mortality rates); northern Sweden, 78 cases of first-ever myocardial
the latency time was 10 years or more (SMR 1.3, 95% infarction were compared with 156 controls. High
CI 1.0-1.5) (Barregård et al., 1990). A similar study from erythrocyte mercury levels or high plasma polyunsat-
Norway comprising 674 male workers from two chlo- urated fatty acids were associated with a low risk of
ralkali plants did not observe increased cardiovascular myocardial infarction (Hallgren et al., 2001).
mortality (Ellingsen et al., 1993). An American study of A case-control study from northern Sweden com-
2133 male workers exposed to mercury vapor during prised 431 cases with myocardial infarction and 499
the production of nuclear weapons did not experience controls. Mercury was measured in erythrocytes: the
increased mortality due to vascular lesions of the cen- median level was 3.54 μg/L. An increased level of mer-
tral nervous system (Cragle et al., 1984). Other vascu- cury in erythrocytes was associated with a lower risk
lar diseases were not reported. of myocardial infarction. The authors propose that
No increase in cardiovascular mortality was mercury levels are a biomarker of fish intake and con-
observed among workers compensated for mercury sider the results to indicate a protective effect of fish
intoxication following exposure in the Italian fur hat consumption (Wennberg et al., 2011).
industry. The number of compensated women and In 1995 the first results were presented from the
men was 820 and 326, respectively (Merler et al., 1994). Kuopio Ischaemic Heart Disease Risk Factor Study
Mortality was studied in 6784 male workers from (KIHD) regarding a possible relation between mer-
four mercury mines and mills in Italy, Slovenia, Spain, cury and heart disease. A total of 1833 men were fol-
and the Ukraine. Increased mortality was found from lowed from 1984 to 1991. Mercury was measured in
hypertension (SMR 1.46, 95% CI 1.08-1.93), heart dis- hair, and men in the highest tertile (> 2.0 μg/g) had
ease other than ischemic (SMR 1.36, 95% CI 1.20-1.53), an increased risk of acute myocardial infarction (RR
and pneumoconiosis (SMR 27.1, 95% CI 23.1-31.6) 2.0, 95% CI 1.2-3.1) and cardiovascular deaths (RR
(­Boffetta et al., 2001). Workers in the mines had the high- 2.9, 95% CI% 1.2-6.6) compared to those with a lower
est SMR for diseases of the heart other than ischemic mercury content (Salonen et al., 1995). The study in
and they were probably exposed to the highest levels eastern Finland was slightly enlarged to comprise
of silica. Cor pulmonale is one of the diagnoses under 1871 men who were followed from 1984 to 1997. A
the category “other diseases of the heart,” and this 44% reduction of acute coronary events was observed
disease may develop as a consequence of pneumoco- when the highest fifth of the serum n-3 end-product
niosis. There were no significant trends between dura- fatty acids docosahexaenoic acid (DHA) and docosa-
tion of employment or estimated cumulative exposure pentaenoic acid (DPA) was compared with the lowest
based on mercury urine levels and hypertension and fifth of these fatty acids. Men in the highest fifth of
16  Cardiovascular Disease 325

the DHA + DPA who had a low hair content of mer- endothelial cell damage by promoting inflamma-
cury had a 67% reduced risk of acute coronary events tion and vasoconstriction as well as lipid peroxida-
(Rissanen et al., 2000). This cohort was followed fur- tion via generation of reactive oxygen species (Grotto
ther until 2002, and now men in the highest third of et al., 2009). Rats were exposed to 100 μg/kg of meth-
hair mercury content (> 2.03 μg/g) had an increased ylmercury chloride per day for 100 days. The long-
risk of acute coronary events (RR 1.60, 95% CI 1.24- term treatment increased the systolic blood pressure.
2.06), CVD (RR 1.68, 95% CI 1.15-2.44) and CHD (RR The treated group also had a decreased level of glu-
1.56, 95% CI 0.99-2.46). A high mercury content in hair tathione in the erythrocytes and an increased level of
attenuated the protective effects of high serum levels malondialdehyde (MDA) in plasma. There was a nega-
of DHA and DPA (Virtanen et al., 2005). A concentra- tive correlation between plasma nitrite/nitrate levels
tion of 2 μg mercury per gram hair corresponds to and systolic blood pressure and a positive correlation
an erythrocyte concentration of 15 ng/g erythrocyte. between MDA and systolic blood pressure, suggest-
This level was exceeded by only two subjects in the ing increased inhibition of NO formation with increase
Swedish study by Hallgren et al. (2001). The discrep- of hypertension (Grotto et al., 2009). This study sug-
ancy between the results of the Finnish and Swedish gests that long-term exposure to methylmercury
studies may be explained by the higher dose in the increases systolic blood pressure, associated at least in
Finnish study. part with increased production of radical oxygen spe-
A case-control study conducted in eight European cies as judged by increased production of MDA and
countries and Israel comprised 684 men with first depressed NO availability (Grotto et al., 2009).
myocardial infarction and 724 referents. Toenail mer- Mercury exposure and oxidative stress were stud-
cury levels were 0.27 μg/g among cases and 0.25 μg/g ied among fish-eating riparians living in the Brazilian
among referents. The risk-factor-adjusted odds ratio Amazon. Both mercury in whole blood and in hair
for myocardial infarction was 2.16 (95% CI 1.09-4.29) were significantly inversely related with several bio-
when the highest quintile of toenail mercury level markers of oxidative stress such as glutathione peroxi-
(0.66 μg/g) was compared with lowest quintile. DHA dase, glutathione, catalase, and δ-aminolevulinic acid
levels in adipose tissue were also included in this dehydratase (ALADH). These results further support
adjustment (Guallar et al., 2002). a relation between mercury exposure and oxidative
Three out of four studies supported a link between stress (Grotto et al., 2010).
methylmercury exposure and intima-media thicken- In summary, the results from studies of workers
ing of the carotid artery (Roman et al., 2011). In one occupationally exposed to mercury are not consistent
of the studies, comprising men from eastern Finland, regarding the risk of CVD. Several studies indicate a
there was a linear relationship between their hair mer- positive association between methylmercury exposure
cury concentration and plasma fibrinogen (Salonen from fish consumption and myocardial infarction. The
et al., 2000), indicating a possible weak inflammatory adverse effect of methylmercury fish intake may be
response. counteracted by the intake of fish-derived fatty acids
Blood pressure was studied among Nunavik Inuit with a protective effect on heart disease.
adults from northern Quebec, Canada. The mean age of
the 732 participants was 34.4 years and the mean blood
mercury level was 50.2 nmol/L (10 μg/L). Mercury was 13 NICKEL
significantly associated with systolic blood pressure
(Valera et al., 2009). Blood pressure was also investi- Humans may be exposed to both metallic nickel
gated in a cross-sectional study of 1240 U.S. women in and nickel compounds. Nickel is a strong skin sensi-
the National Health and Nutrition Examination Sur- tizer (see Chapter 48 “Nickel” of this book), and nickel
vey (NHANES) 1999-2000. The mean concentration of compounds have been classified as human carcinogen
mercury in blood was 1.8 μg/L. Fish consumers had by the International Agency for Research on Cancer
a higher mean concentration compared with nonfish (IARC, 2012). The IARC noted that the carcinogenic
consumers: 2.3 and 0.8 μg/L, respectively. A significant action of nickel compounds may involve an inflam-
relationship was found between blood mercury levels matory process, and a specific chronic inflammation
and systolic blood pressure among nonfish consum- mechanism at the cellular level has been identified
ers but not among fish consumers. The intake of fish after inhalation of nickel compounds in experimental
oils may counteract the harmful effects of mercury on animals (Goodman et al., 2011).
blood pressure (Vupputuri et al., 2005). Most studies of nickel-exposed workers focus on
Methylmercury exposure can cause oxidative stress, skin sensitization and cancer, and only a few have
an early biological response that can produce vascular reported mortality from CVD.
326 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

No increase in circulatory diseases was reported suspected as part of the etiology, and selenium defi-
in 812 nickel carbonyl refinery workers in Wales, fol- ciency is currently considered to be a conditioning pre-
lowed for mortality between 1958 and 2000. There were disposing (rather than a causative) factor for Keshan
85 deaths from circulatory diseases (SMR 0.94, 95% CI disease (Tanguy et al., 2012). Cardiomyopathy has also
0.75-1.16) (Sorahan and Williams, 2005). There was a been described in selenium deficiency resulting from
deficit in deaths from circulatory diseases in a cohort prolonged parenteral alimentation (Johnson et al.,
of 1649 male workers at a nickel refinery and fertilizer 1981). In a prospective cohort study conducted in Fin-
plant in Fort Saskatchewan, Alberta, Canada. During land, a country with a low selenium intake, Salonen
follow-up (1954-1995), the SMR for circulatory dis- and coworkers (1982) found a strong inverse associa-
eases was 0.57 (95% CI 0.36-0.87), based on 22 deaths tion between selenium and cardiovascular death and
(Egedahl et al., 2001). Lightfoot et al. (2010) reported myocardial infarction in a matched-pair longitudinal
mortality findings in a large cohort of 10253 male nickel study. Since then, several cohort studies have evalu-
and copper production workers in Sudbury, Ontario, ated the association between selenium biomarkers and
Canada. The number of deaths from circulatory dis- coronary risk, with inconsistent findings (Navas-Acien
eases during 1964-2001 was close to the expected num- et al., 2008b). Randomized controlled trials have not
ber for both the first 15 years after employment (SMR detected any major effects of selenium supplementa-
0.96, 95% CI 0.72-1.24) and after 15 years or more (SMR tion on CVD incidence (Hercberg et al., 2004; Stranges
0.99, 95% CI 0.92-1.06). et al., 2006). Selenium was not significantly associated
Enterline and Marsh (1982) reported mortality with any of the CVD endpoints (all CVD, myocardial
among nickel refinery workers in West Virginina, USA. infarction, stroke, all-CVD mortality) during 7.6 years
The cohort comprised 1865 workers hired before 1947 of follow-up, even after analyses were further stratified
and 1354 workers hired after 1946. A statistically insig- by tertiles of baseline plasma selenium concentrations
nificant excess incidence of stroke was noted among (Stranges et al., 2006). In a meta-analysis of six stud-
those employed before 1947 (SMR 1.62, 12 cases). ies, the pooled RR for CHD endpoints of supplements
No excess stroke incidence was noted among those containing selenium compared with placebo was 0.89
employed after 1946, and there was a deficit of deaths (95% CI 0.68-1.17). The authors concluded that the
due to heart disease in both subcohorts. evidence was inadequate to establish a protective role
In summary, only a few studies of nickel-exposed for selenium supplements against CHD (Flores-Mateo
workers have reported mortality from CVD. There et al., 2006).
is very little evidence of an increased risk of CVD in A meta-analysis of observational (14 prospective
these studies. It should be noted, though, that the stud- cohort studies and 11 case-control, most of them con-
ies have mainly focused on evaluating cancer risks and ducted in Europe) studies on serum or plasma sele-
there has been less emphasis on exploring a potential nium levels and CHD was performed by Flores-Mateo
association with CVD. and coworkers (2006). The pooled RR, when compar-
ing the highest to the lowest category of selenium con-
centration, was 0.85 in cohort studies (95% CI 0.74-0.99)
14 SELENIUM and 0.43 in case-control studies (95% CI 0.29-0.66). The
authors conclude that the validity of the association
Selenium is an essential trace element (although is, however, uncertain because observational studies
toxic in large doses) necessary for the activity of many have been unreliable in determining the cardiovascu-
enzymes and proteins; it is derived mainly from plant lar effects of other antioxidants and vitamins, and that,
foods, animal proteins, and seafood. Occupational overall, the evidence is still inadequate to establish a
exposure may occur in metal industries, selenium- protective role for selenium in CHD.
recovery processes, and painting and special trades. Navas-Acien and coworkers (2008b) reviewed the
It has been hypothesized that the antioxidant prop- role of selenium in the prevention of atherosclerotic
erties of selenium, especially on the glutathione-­ CVD. The authors concluded that current evidence is
peroxidase-system, and decreased lipid peroxidation insufficient to support a protective role for selenium in
may play an important role in the prevention of CVD prevention. They also stated that data from trials
atherosclerotic CVD. of selenium-containing supplements and from epide-
Keshan disease (dilated cardiomyopathy) has been miological studies suggest that chronically increased
linked to selenium deficiency, mainly in children and selenium intake in selenium-replete populations can
young women from the Keshan region of China; it induce diabetes and maybe also hypercholesterolemia.
is often reversible after selenium supplementation. A recent review by Tanguy and coworkers (2012) pres-
However, a role for an infectious agent has also been ents the current knowledge on the role of selenium in
16  Cardiovascular Disease 327

cardiac health and describes growing epidemiologi- Kreuzer et al. (2010) reported a cohort study of 58,987
cal evidence of several side effects of high selenium male German uranium miners who were followed for
intake that may negatively influence cardiovascular mortality from 1946 to 2003. A cohort-internal analy-
health. Recent studies have shown that very moder- sis relating radiation dose in terms of working level
ate increases in selenium status, far below doses that months (WLM) to cause-specific mortality showed no
induce intoxication symptoms (nausea, vomiting, nail evidence of an association with CHD or cerebrovascu-
discoloration, hair loss, fatigue, and orthostatic hypo- lar disease.
tension), can increase cardiometabolic risk factors such Canu et al. (2012) reported a cohort mortality study
as T2DM or dyslipidemia in populations with a nor- of workers at a French uranium-enrichment plant.
mal selenium intake (Tanguy et al., 2012). A total of 2897 workers (of which 188 were women)
In summary, selenium deficiency has been related employed between 1960 and 2006 were followed for
to dilated cardiomyopathy. Selenium may have a pro- mortality from 1968 to 2007. Exposure was classified in
tective role against CVD, but the evidence is insuf- terms of exposure to natural and reprocessed uranium,
ficient. There are also data suggesting that increased and in terms of solubility (fast, medium, or slow), by
selenium intake may increase the risk of diabetes and a job exposure matrix. An increased risk of death from
dyslipidemia. CVD in general was observed, particularly in associa-
tion with slow solubility reprocessed uranium, which
is thought to be associated with a higher internal irra-
15 URANIUM diation dose. The number of deaths from myocardial
infarction and cerebrovascular disease was small, pre-
Natural uranium is composed of several isotopes cluding conclusions regarding the relation to uranium
and may exert negative health effects both from tox- exposure.
icity of the metal and from the associated ionizing No statistically significant association was found
irradiation. between urinary levels of uranium and CHD or stroke
It is well known that high exposure to ionizing irra- in a cross-sectional study of 1857 U.S. adults in the
diation causes an increased risk of developing CVDs. NHANES (Mendy et al., 2012).
Irradiation therapy for cancer of the thorax is associ- In summary, high exposure to ionizing irradiation
ated with an increased risk of myocardial infarction, such as in patients treated with radiotherapy and in
and patients treated with irradiation for tumors of atomic bomb survivors is causally linked to an excess
the head and neck are at an increased risk of cere- of CVD. Studies in uranium miners and uranium pro-
brovascular diseases (Travis et al., 2012). Studies of cessing workers show less consistent evidence but sug-
atomic bomb survivors have shown an elevated risk gest an excess of stroke (in particular) in association
of both CVDs and cerebrovascular diseases (Preston with exposure to ionizing irradiation from uranium.
et al., 2012).
Studies of occupational exposure to ionizing irra-
diation show diverging results. Excess mortality from 16 ZINC
stroke has been reported among nuclear power plant
workers (Azizova et al., 2011; Laurent et al., 2010), Zinc was established to be an essential trace metal
Chernobyl emergency workers (Ivanov, 2007), and in 1961 following the discovery of zinc deficiency in
medical X-ray technicians (Hauptmann et al., 2003). humans. Zinc is normally absorbed from ingested
No excess of stroke or circulatory diseases was found food. It is one of the most abundant elements within
in a multicenter study of nuclear power plant workers, cells and is necessary for a broad range of physiologi-
although statistical power may have been low among cal processes. An initial consequence of zinc deficiency
highly exposed workers (Vrijheid et al., 2007). is impaired immunological functions. Zinc deficiency
There are a few studies in workers specifically is reported to contribute significantly to the global bur-
exposed to uranium. Nusinovici et al. (2010) reported den of disease (Foster and Samman, 2012). Zinc fume
a study of mortality (during 1946-1999) in a cohort of fever is a well-known effect of inhalation of newly gen-
5085 male French uranium miners. Exposure was char- erated fumes (Hunter’s diseases of occupation, 2010).
acterized based on dosimetry. The RR of stroke among A meta-analysis of 33 controlled clinical trials and
exposed versus unexposed subjects in the cohort was 14,238 subjects was conducted to determine the effect
1.39 (95% CI 0.81-2.38), and there was a positive dose- of zinc supplementation on plasma cholesterol and
response relationship (P = 0.03). There was no indica- triglyceride concentrations in humans. Plasma zinc
tion of an association between exposure to uranium concentrations increased significantly. No effect of zinc
and IHD. supplementation on plasma lipoprotein was detected
328 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

in the overall analysis. In individuals classified as Cardiomyopathy has been associated with exces-
healthy, zinc supplementation was associated with a sive intake of arsenic, as well as cobalt, in beer. Iron
decrease in high-density lipoprotein cholesterol levels, overload cardiomyopathy is a consequence of iron
thus contributing to an increased risk of CHD (Foster accumulation in the myocardium, mainly caused by
et al., 2010). However, in a study of patients with T2DM genetically determined disorders of iron metabolism
a higher serum concentration of zinc was associated or multiple transfusions. Deficiency of selenium has
with a decreased risk of myocardial infarction (Soinio been linked to dilated cardiomyopathy in children and
et al., 2007). Oral zinc sulfate does not appear to aid the young women from the Keshan region of China.
healing of arterial and venous ulcers (Wilkinson, 2012). Cor pulmonale has been associated with airborne
Inhalation of 2.5 or 5 mg/m3 zinc oxide fume for 2 h in exposure to beryllium, and signs of this disease have
human volunteers increased the plasma concentration also been observed after cobalt exposure among hard-
of IL-6. This level of exposure also resulted in mild fever metal workers. Pulmonary fibrosis with increased
(Fine et al., 1997). In a later study, sheet-metal workers blood pressure in the pulmonary circulation is a likely
occupationally exposed to low levels of zinc oxide were mechanism in the development of cor pulmonale.
exposed to 5 mg/m3. Despite a mild clinical response, Various types of systematic errors may introduce
the mean plasma IL-6 levels increased significantly after bias in epidemiological studies. One type of systemic
exposure. The inflammatory response decreased in naive error is inadequate comparability between exposed
subjects exposed on three successive days (Fine et al., workers and nonexposed referents. In many historical
2000). These experiments indicate adaptation, with a occupational cohort studies, disease or death rates of
milder inflammatory response after repeated exposures. exposed workers are compared with those of the gen-
Welders are sometimes exposed to zinc oxide fumes eral national or regional population. Such comparisons
when welding galvanized steel. In a survey, 31% of are likely to result in underestimation of the true risk
welders aged between 20 and 59 years had experi- as the general population includes sick and disabled
enced metal fume fever on at least one occasion (Ross, people who are unable to work. This underestimation
1974). However, not all metal fume fevers are caused is known as the healthy worker effect (McMichael, 1976;
by zinc oxide. Baillargeon, 2001). This bias may explain the decreased
The California Teachers Study comprised approxi- risk reported in many occupational cohort studies
mately 45,000 women. Long-term airborne exposure focusing on CVDs. Consequently, results may have
to PM2.5 doubled mortality from IHD. Airborne zinc been incorrectly interpreted as showing that no rela-
exposure correlated with PM2.5 exposure and had an tionship exists between a specific exposure and CVD.
effect which was much weaker (Ostro et al., 2010). Since the late twentieth century, particulate envi-
In summary, the impact of zinc supplementation or ronmental air pollutants have attracted increasing
serum levels of zinc on the occurrence of CVD cannot interest. In 2010 the American Heart Association
be evaluated because long-term occupational cohort summarized the epidemiological evidence for the
studies have not been done. cardiovascular effects of PM2.5, traffic-related or com-
bustion-related, air pollution exposure at ambient
levels. There was strong overall epidemiological evi-
17  CONCLUDING REMARKS dence for an association between short-term exposure
(days) or longer-term exposure (months to years) and
There is strong epidemiological and mechanistic IHD (Brook et al., 2010). The American Heart Asso-
evidence for an association between exposure to arse- ciation also stated strong overall mechanistic evidence
nic and lead and the development of CVD. There is rel- of a relation between a systemic proinflammatory
atively strong but less conclusive evidence for a causal response and cardiovascular effects in humans as well
relationship between cadmium and mercury exposure as in animals (Brook et al., 2010). The theory linking
and CVD. All of these metals are suspected of inducing environmental and occupational air pollutants with
pathophysiological changes relevant to atherogenic IHD via low-grade inflammation was launched in the
disorders, including increased oxidative stress, inflam- mid-1990s (Seaton et al., 1995; Sjögren, 1997). There is
matory response, and coagulation activity (Alissa and some evidence that inhalation of metal particles may
Ferns, 2011). induce an inflammatory response associated with an
It is quite well established that high exposure to increased blood coagulation. Inhalation of zinc or
ionizing irradiation increases the risk for both CVDs copper may cause metal fume fever (Hunter’s dis-
and cerebrovascular diseases. Irradiation may explain eases of occupation, 2010). Experimental exposure to
the suggested relation between uranium exposure and zinc oxide increased the serum level of IL-6, which is
cerebrovascular disease. a marker of inflammation (Fine et al., 1997) and a risk
16  Cardiovascular Disease 329

factor for CHD (Danesh et al., 2008). Welding fume Ellingsen, D.G., Andersen, A., Nordhagen, H.P., et al., 1993. Br. J. Ind.
exposure increased another inflammatory marker, Med. 50, 875–880.
Enterline, P.E., 1974. J. Occup. Med. 16, 523–526.
CRP, which is also a risk factor for CHD (Danesh et al., Enterline, P.E., Marsh, G.M., 1982. J. Natl. Cancer Inst. 68, 925–933.
1998). In some studies focusing on environmental par- Epstein, S.E., Zhou, Y.F., Zhu, J., 1999. Circulation 100, e20–e28.
ticulate air pollutants, some single metals (e.g. nickel) Fagerberg, B., Bergström, G., Borén, J., et al., 2012. J. Intern. Med.
have shown relations with CVD (Lippmann et al., 272, 601–610.
2006; Mostofsky et al., 2012; Ostro et al., 2010; WHO, Fagerberg, B., Bergström, G., Borén, J., et al., 2013. BMJ Open. Mar 6
3 (3). pii: e002489. doi: 10.1136/bmjopen-2012-002489.
2013). These studies must be interpreted with caution Fernandes, J.L., Storey, P., da Silva, J.A., et al., 2011. J. Cardiovasc.
as these results are based on multiple comparisons Magn. Reson 13, 6.
with a significant result for only one or two metals. Fine, J.M., Gordon, T., Chen, L.C., et al., 1997. J. Occup. Environ.
Med. 39, 722–726.
Fine, J.M., Gordon, T., Chen, L.C., et al., 2000. J. Occup. Environ.
Med. 42, 1085–1091.
References Flores-Mateo, G., Navas-Acien, A., Pastor-Barriuso, R., et al., 2006.
Am. J. Clin. Nutr. 84, 762–773.
Abhyankar, L.N., Jones, M.R., Guallar, E., et al., 2012. Environ. Flynn, M.R., Susi, P., 2010. J. Occup. Environ. Hyg. 7, 115–126.
Health. Perspect. 120, 494–500. Foster, M., Samman, S., 2012. Nutrients 4, 676–694.
Agarwal, S., Zaman, T., Tuzcu, E.M., et al., 2011. Angiology 62, 422–429. Foster, M., Petocz, P., Samman, S., 2010. Atherosclerosis 210, 344–352.
Alexander, C.S., 1969. Editorial. Ann. Intern. Med. 70, 411–413. Friesen, M.C., Demers, P.A., Spinelli, J.J., et al., 2010. Am. J. Epide-
Alissa, E.M., Ferns, G.A., 2011. J. Toxicol. 2011, 870125. miol. 172, 790–799.
Ascherio, A., Rimm, E.B., Hernán, M.A., et al., 1998. Circulation 98, Frostegård, J., 2010. Arterioscler. Thromb. Vasc. Biol. 30, 892–893.
1198–1204. Gallagher, C.M., Meliker, J.R., 2010. Environ. Health Perspect. 118,
Axelson, O., Dahlgren, E., Jansson, C.-D., et al., 1978. Brit. J. Ind. 1676–1684.
Med. 35, 8–15. Gerstner, H.B., Huff, J.E., 1977. J. Toxicol. Environ. Health 2, 491–526.
Azizova, T.V., Muirhead, C.R., Moseeva, M.B., et al., 2011. Radiat. Goldsmith, S., From, A.H.L., 1980. N. Engl. J. Med. 303, 1096–1098.
Environ. Biophys. 50, 539–552. Goodman, J.E., Prueitt, R.L., Thakali, S., et al., 2011. Crit. Rev. Toxicol.
Baillargeon, J., 2001. Occup. Med. 16, 359–366. 41, 142–174.
Barregård, L., Sällsten, G., Järvholm, B., 1990. Br. J. Ind. Med. 47, 99–104. Grotto, D., de Castro, M.M., Barcelos, G.R.M., et al., 2009. Arch.
Beaumont, J.J., Weiss, N.S., 1980. Am. J. Epidemiol. 112, 775–786. ­Toxicol. 83, 653–662.
Becker, N., 1999. J. Occup. Environ. Med. 41, 294–303. Grotto, D., Valentini, J., Fillion, M., et al., 2010. Sci. Total Environ.
Binks, K., Doll, R., Gillies, M., et al., 2005. Occup. Med. (Lond) 55, 408, 806–811.
215–226. Guallar, E., Sanz-Gallardo, M.I., van´t Veer, P., et al., 2002. N. Engl. J.
Birk, T., Mundt, K.A., Dell, L.D., et al., 2006. J. Occup. Environ. Med. Med. 347, 1747–1754.
48, 426–433. Hallgren, C.G., Hallmans, G., Jansson, J.-H., et al., 2001. Brit. J. Nutr.
Boffetta, P., Sällsten, G., Garcia-Gomez, M., et al., 2001. Occup. 86, 397–404.
­Environ. Med. 58, 461–466. Hartmann, A.L., Hartmann, W., Buhlmann, A.A., 1983. Schweiz.
Brook, R.D., Rajagopalan, S., Pope 3rd, C.A., et al., 2010. Circulation Med. Wochenschr 113, 766–770.
121, 2331–2378. Hauptmann, M., Mohan, A.K., Doody, M.M., et al., 2003. Am. J. Epi-
Canu, I.G., Garsi, J.-P., Caer-Lorho, S., et al., 2012. Occup. Environ. demiol. 157, 239–248.
Med. 69, 404–409. Hayes, R.B., Sheffet, A., Spirtas, R., 1989. Am. J. Ind. Med. 16, 127–133.
Cowan, D.M., Zheng, W., Zou, Y., et al., 2009. Neurotoxicol 30, Hercberg, S., Galan, P., Preziosi, P., et al., 2004. Arch. Intern. Med.
1214–1222. 164, 2335–2342.
Cragle, D.L., Hollis, D.R., Qualters, J.R., et al., 1984. J. Occup. Med. Hertz-Picciotto, I., Arrighi, H.M., Hu, S.-W., 2000. Am. J. Epidemiol.
26, 817–821. 151, 174–181.
Crump, C., Crump, K., Hack, E., et al., 2003. Risk Anal. 23, 1147–1163. Hobbesland, Å., Kjuus, H., Thelle, D.S., 1997a. Scand. J. Work
Danesh, J., Collins, R., Appleby, P., et al., 1998. J. A. M. A. 279, 1477–1482. ­Environ. Health. 23, 342–350.
Danesh, J., Whincup, P., Walker, M., et al., 2000. B. M. J. 321, 199–204. Hobbesland, Å., Kjuus, H., Thelle, D.S., 1997b. Scand. J. Work
Danesh, J., Lewington, S., Thompson, S.G., et al., 2005. J. A. M. A. ­Environ. Health. 23, 334–341.
294, 1799–1809. Hogstedt, C., Alexandersson, R., 1990. Arbete och Hälsa 1990, 21
Danesh, J., Kaptoge, S., Mann, A.,G., et al., 2008. PLoS Medicine 5 (In Swedish with English abstract).
(e78), 0600–0610. Horowitz, S.F., Fischbein, A., Matza, D., et al., 1988. Br. J. Ind. Med.
Davies, J.M., Easton, D.F., Bidstrup, P.L., 1991. Br. J. Ind. Med. 48, 45, 742–746.
299–313. Houston, M., 2011. J. Clin. Hypertens 13, 843–847.
Dickinson, H.O., Nicolson, D.J., Campbell, F., et al., 2006. Cochrane Hunter, D., 1978. The diseases of occupation, sixth ed. Hodder and
Database Syst. Rev. Jul. 19. CD004640. Stoughton Ltd, Kent, Great Britain; p. 1257.
Dickinson, H.O., Nicolson, D.J., Campbell, F., et al., 2009. Cochrane Hunter’s diseases of occupation, 2010. In: Baxter, P.J., Aw, T.-C.,
Library 2009; Issue 1. http://www.thecochranelibrary.com/ Cockcroft, A., Harrington, J.M. (Eds.), tenth ed. Hodder and
view/0/index.html (accessed 10.12.13). Stoughton Ltd, London, U.K.
Dorhout Mees, S.M., Algra, A., Vandertop, W.P., et al., 2012. Lancet IARC (International Agency for Research on Cancer), 2012. IARC
380, 44–49. Monographs on the Evaluation of Carcinogenic Risks to H ­ umans.
Egedahl, R., Carpenter, M., Lundell, D., 2001. Occup. Environ. Med. A Review of Human Carcinogens: Arsenic, Metals, Fibres, and
58, 711–715. Dusts, Vol. 100C. International Agency for Research on Cancer
Elin, R.J., 1987. Clin. Chem. 33, 1965–1970. Lyon, France.
330 Bengt Sjögren, Carolina Bigert, and Per Gustavsson

Ibfelt, E., Bonde, J.P., Hansen, J., 2010. Occup. Environ. Med. 67, Mendy, A., Gasana, J., Vieira, E.R., 2012. Int. J. Environ. Health Res.
772–777. 22, 105–118.
Ilychova, S.A., Zaridze, D.G., 2012. Occup. Environ. Med. 69, 87–92. Merler, E., Boffetta, P., Masala, G., et al., 1994. J. Occup. Med 36,
ISIS-4, 1995. Lancet 345, 669–685. 1260–1264.
Iso, H., Stampfer, M.J., Manson, J.E., et al., 1999. Stroke 30, 1772–1779. Michaels, D., Zoloth, S.R., Stern, F.B., 1991. Int. J. Epidemiol. 20,
Ivanov, V.K., 2007. Health Phys. 93, 470–479. 978–983.
Järup, L., Pershagen, G., Wall, S., 1989. Am. J. Ind. Med. 15, 31–41. Moon, K., Guallar, E., Navas-Acien, A., 2012. Curr. Atheroscler. Rep.
Järup, L., Bellander, T., Hogstedt, C., et al., 1998. Occup. Environ. 14 (6), 542–555.
Med. 55, 755–759. Mordukhovich, I., Wright, R.O., Hu, H., et al., 2012. Environ. Health
Jarvis, J.Q., Hammond, E., Meier, R., et al., 1992. J. Occup. Med. 34, Perspect 120, 98–104.
620–626. Morin, Y., Tetu, A., Mercier, G., 1971. Br. Heart J. 33, 175–178. supple-
Jee, S.A., Miller III., E.R., Guallar, E., et al., 2002. Am. J. Hypertens. ment.
15, 691–696. Mostofsky, E., Schwartz, J., Coull, B.A., et al., 2012. Am. J. Epidemiol.
Johnson, R.A., Baker, S.S., Fallon, J.T., et al., 1981. N. Engl. J. Med. 176, 317–326.
304, 1210–1212. Moulin, J.J., Wild, P., Haguenoer, J.M., et al., 1993. Br. J. Ind. Med.
Kalliomäki, P.-L., Alanko, K., Korhonen, O., et al., 1978. Scand. J. 50, 234–243.
Work Environ. Health 4, 122–130. Moulin, J.J., Wild, P., Romazini, S., et al., 1998. Am. J. Epidemiol. 148,
Kang, P., Liu, T., Tian, C., et al., 2012. Clin. Chim. Acta. 413, 241–248.
1424–1429. Moyer, C.F., Kodavanti, U.P., Haseman, J.K., et al., 2002. Toxicol.
Kazantzis, G., Lam, T.H., Sullivan, K.R., 1988. Scand. J. Work. Envi- Pathol. 30, 427–434.
ron. Health 14, 220–223. Mozaffarian, D., Shi, P., Morris, J.S., et al., 2011. N. Engl. J. Med. 364,
Kennedy, A., Dornan, J.D., King, R., 1981. Lancet 317, 412–414. 1116–1125.
Kim, J.Y., Chen, J.-C., Boyce, P.D., et al., 2005. Occup. Environ. Med. Nakagawa, H., Nishijo, M., Morikawa, Y., et al., 2006. Environ. Res.
62, 157–163. 100, 323–329.
Klatsky, A.L., 2006. Am. J. Epidemiol. 164, 194–195. Nakanishi, S., Yamane, K., Ohishi, W., et al., 2008. Diab. Res. Clin.
Korallus, U., Ulm, K., Steinmann-Steiner-Haldenstaett, W., 1993. Int. Pract. 81, 381–385.
Arch. Occup. Environ. Health 65, 171–178. NTP (National Toxicology Program), 2012. NTP Monograph on
Kremastinos, D.T., Farmakis, D., 2011. Circulation 124, 2253–2263. Health Effects of Low-Level Lead. U.S. Department of Health
Kreuzer, M., Grosche, B., Schnelzer, M., et al., 2010. Radiat. Environ. and Human Services. http://ntp.niehs.nih.gov/NTP/ohat/
Biophys. 49, 177–185. Lead/Final/MonographHealthEffectsLowLevelLead_NewISSN_
Kucharska-Newton, A.M., Couper, D.J., Pankow, J.S., et al., 2009. 508.pdf (accessed 10.12.13).
­Arterioscler. Thromb. Vasc. Biol. 29, 2182–2190. Navas-Acien, A., Selvin, E., Sharrett, A.R., et al., 2004. Circulation
Laurent, O., Metz-Flamant, C., Rogel, A., et al., 2010. Intl. Arch. 109, 3196–3201.
­Occup. Environ. Health 83, 935–944. Navas-Acien, A., Silbergeld, E.K., Sharrett, R., et al., 2005. Environ.
Lauwerys, R., Lison, D., 1994. Sci. Total Environ. 150, 1–6. Health Perspect. 113, 164–169.
Lee, A.M., Fraumeni Jr, J.F., 1969. J. Natl. Cancer. Inst. 42, 1045–1052. Navas-Acien, A., Guallar, E., Silbergeld, E.K., et al., 2007. Environ.
Lee, B.-K., Kim, Y., 2011. Environ. Res. 111. 7978–7803. Health Perspect. 115, 472–482.
Lee-Feldstein, A., 1983. J. Natl. Cancer Inst. 70, 601–610. Navas-Acien, A., Bleys, J., Guallar, E., 2008b. Curr. Opin. Lipidol. 19,
Li, J., Zhang, Q., Zhang, M., et al., 2007. Cochrane Database Syst. 43–49.
Rev.. Issue 2 Art No. CD002755. Navas-Acien, A., Schwartz, B.S., Rothenberg, S.J., et al., 2008a.
Li, J., Zhang, Q., Zhang, M., et al., 2009. Cochrane Library Issue 1. ­Epidemiology 19, 496–504.
http://onlinelibrary.wiley.com/doi/10.1002/14651858.CD002755. Nawrot, T.S., Thijs, L., Den Hond, E.M., et al., 2002. J. Hum. Hypertens.
pub2/pdf/standard (accessed 10.12.13). 16, 123–131.
Liao, F., Folsom, A.R., Brancati, F.L., 1998. Am. Heart J. 136, Nawrot, T.S., Van Hecke, E., Thijs, L., et al., 2008. Environ. Health
480–490. Perspect. 116, 1620–1628.
Libby, P., Ridker, P.M., Maseri, A., 2002. Circulation 105, 1135–1143. Newhouse, M.L., Oakes, D., Woolley, A.J., 1985. Br. J. Ind. Med. 42,
Lightfoot, N., Berriault, C., Semenciw, R., 2010. Occup. Med. (Lond) 406–410.
60, 211–218. Nishijo, M., Morikawa, Y., Nakagawa, H., et al., 2006. Occup. Envi-
Lim, S.S., Vos, T., Flaxman, A.D., et al., 2012. Lancet 380, 2224–2260. ron. Med. 63, 545–550.
Lippmann, M., Ito, K., Hwang, J.-S., et al., 2006. Environ. Health Nusinovici, S., Vacquier, B., Leuraud, K., et al., 2010. Scand. J. Work
­Perspect. 114, 1662–1669. Environ. Health 36, 373–383.
Lu, L., Zhang, L.-I., Li, G.J., et al., 2005. Neurotoxicol 26, 257–265. Ohira, T., Peacock, J.M., Iso, H., et al., 2009. Am. J. Epidemiol. 169,
Luippold, R.S., Mundt, K.A., Austin, R.P., et al., 2003. Occup. Envi- 1437–1444.
ron. Med. 60, 451–457. Ostro, B., Lipsett, M., Reynolds, P., et al., 2010. Environ. Health Per-
Luippold, R.S., Mundt, K.A., Dell, L.D., et al., 2005. J. Occup. Envi- spect. 118, 363–369.
ron. Med. 47, 381–385. Pastides, H., Austin, R., Lemeshow, S., et al., 1994. Am. J. Ind. Med.
Manna, P., Sinha, M., Sil, P.C., 2008. Arch. Toxicol. 82, 137–149. 25, 663–675.
Marsh, G.M., Esmen, N.A., Buchanich, J.M., et al., 2009. Am. J. Ind. Peacock, J.M., Folsom, A.R., Arnett, D.K., et al., 1999. Ann. Epide-
Med. 52, 633–644. miol. 9, 159–165.
Marx, A., Neutra, R.R., 1997. Epidemiol. Rev. 19, 258–272. Peters, J.L., Perlstein, T.S., Perry, M.J., et al., 2010. Environ. Res. 110,
Mayerl, C., Lukasser, M., Sedivy, R., et al., 2006. Virchows Arch. 449, 199–206.
96–103. Peters, S., Reid, A., Fritschi, L., et al., 2013. Occup. Environ. Med. 70
McMichael, A.J., 1976. J. Occup. Med. 18, 165–168. (12), 864–868.
Medrano, M.J., Boix, R., Pastor-Barriuso, R., et al., 2010. Environ. Res. Preston, D.L., Shimizu, Y., Pierce, D.A., et al., 2012. Radiat. Res. 178,
110, 448–454. AV146–AV172.
16  Cardiovascular Disease 331

Ridker, P.M., 1999. Blood. Coagul. Fibrinolysis. 10 (Suppl. 1), S9–S12. Tellez-Plaza, M., Jones, M.R., Dominguez-Lucas, A., et al., 2013b.
Rissanen, T., Voutilainen, S., Nyyssönen, K., et al., 2000. Circulation Curr Atheroscler Rep. 15, 356. http://dx.doi.org/10.1007/
102, 2677–2679. s11883-013-0356-2.
Rodier, J., 1955. Br. J. Ind. Med. 12, 21–35. Thériault, G.P., Tremblay, C.G., Armstrong, B.G., 1988. Am. J. Ind.
Rodríguez-Morán, M., Simental Mendía, L.E., Zambrano Galván, G., Med. 13, 659–666.
et al., 2011. Magnesium Res. 24, 156–162. Travis, L.B., Ng, A.K., Allan, J.M., et al., 2012. J. Natl. Cancer Inst.
Roels, H., Lauwerys, R., Buchet, J.-P., et al., 1987. Am. J. Ind. Med. 104, 357–370.
11, 307–327. Valera, B., Dewailly, E., Poirier, P., 2009. Hypertension 54, 981–986.
Roman, H.A., Walsh, T.L., Coull, B.A., et al., 2011. Environ. Health Virtanen, J.K., Voutilainen, S., Rissanen, T.H., et al., 2005. Arterio-
Perspect. 119, 607–614. scler. Thromb. Vasc. Biol. 25, 228–233.
Rønneberg, A., 1995. Occup. Environ. Med. 52, 255–261. Vrijheid, M., Cardis, E., Ashmore, P., et al., 2007. Int. J. Epidemiol.
Rosenlund, M., Berglind, N., Hallqvist, J., et al., 2005. Epidemiology 36, 1126–1135.
16, 570–576. Vupputuri, S., Longnecker, M.P., Daniels, J.L., et al., 2005. Environ.
Rosenman, K.D., Stanbury, M., 1996. Am. J. Ind. Med. 29, 491–500. Res. 97, 195–200.
Ross, D.S., 1974. J. Soc. Occup. Med 24, 125–129. Wagoner, J.K., Infante, P., Bayliss, D.L., 1980. Environ. Res. 21, 15–34.
Ross, R., 1993. Nature 362, 801–809. Wang, J., Pantopoulos, K., 2011. Biochem. J. 434, 365–381.
Ross, R., 1999. N. Engl. J. Med. 340, 115–126. Ward, E., Okun, A., Ruder, A., et al., 1992. Am. J. Ind. Med. 22,
Ryzen, E., Servis, K.L., DeRusso, P., et al., 1989. J. Am. Coll. Nutr. 8, 885–904.
580–587. Weglicki, W.B., 2012. Annu. Rev. Nutr. 32, 55–71.
Salonen, J.T., Alfthan, G., Huttunen, J.K., et al., 1982. Lancet 2 (8291) Weinbruch, S., Dirsch, T., Ebert, M., et al., 2008. J. Environ. Monit.
(Jul 24), 175–179. 10, 648–654.
Salonen, J.T., Seppänen, K., Nyyssönen, K., et al., 1995. Circulation Weisskopf, M.G., Jain, N., Nie, H., et al., 2009. Circulation 120,
91, 645–655. 1056–1064.
Salonen, J.T., Seppänen, K., Lakka, T.A., et al., 2000. Atherosclerosis Welch, K., Higgins, I., Oh, M., et al., 1982. Arch. Environ. Health 37,
148, 265–273. 325–335.
Saric, M., Hrustic, O., 1975. Environ. Res. 10, 314–318. Wennberg, M., Bergdahl, I.A., Hallmans, G., et al., 2011. Am. J. Clin.
Satoh, K., Fukuda, Y., Torii, K., et al., 1981. J. Occup. Med. 23, 835–838. Nutr. 93, 27–36.
Sawyer, R.,T., Maier, L.A., 2011. Biometals 24, 1–17. Wheeler, J.G., Mussolino, M.E., Gillum, R.F., et al., 2004. Euro. Heart J.
Schubauer-Berigan, M.K., Couch, J.R., Petersen, M.R., et al., 2011. 25, 1287–1292.
Occup. Environ. Med. 68, 345–353. WHO (World Health Organization), 2012. WHO Regional Office for
Seaton, A., MacNee, W., Donaldson, K., et al., 1995. Lancet 345, 176–178. Europe. Definition of cardiovascular diseases. http://www.euro.
Seghizzi, P., D’Adda, F., Borleri, D., et al., 1994. Sci. Total Environ. who.int/en/health-topics/noncommunicable-diseases/
150, 105–109. cardiovascular-diseases/cardiovascular-diseases2/definition-­of-
Simonato, L., Fletcher, A.C., Andersen, A., et al., 1991. Br. J. Ind. Med. cardiovascular-diseases (accessed 10.12.13).
48, 145–154. WHO, 2013. Regional Office for Europe. Review of evidence on health
Sjögren, B., 1997. Occup. Environ. Med. 54, 466–469. aspects of air pollution—REVIHAAP Project: final technical report.
Sjögren, B., Knutsson, A., Bergström, H., et al., 2002. C. Eur. J. Occup. http://www.euro.who.int/en/health-topics/environment-and-
Environ. Med. 8, 49–54. health/air-quality/publications/2013/review-of-evidence-on-
Soinio, M., Marniemi, J., Laakso, M., et al., 2007. Diabetes Care 30, health-aspects-of-air-pollution-revihaap-project-final-­technical-
523–528. report (accessed 10.12.13).
Sorahan, T., Williams, S.P., 2005. Occup. Environ. Med. 62, 80–85. Wiebert, P., Lönn, M., Fremling, K., et al., 2012. Occup. Environ. Med.
Sorahan, T., Lister, A., Gilthorpe, M.S., et al., 1995. Occup. Environ. 69, 651–657.
Med. 52, 804–812. Wilkinson, E.A.J., 2012. Cochrane Database of Systematic Reviews.
Steenland, K., Selevan, S., Landrigan, P., 1992. Am.. J. Public Health http://dx.doi.org/10.1002/14651858.CD001273.pub2. Issue 8.
82, 1641–1644. Art. No.: CD001273.
Stranges, S., Marshall, J.R., Trevisan, M., et al., 2006. Am. J. Epide- Yoshizawa, K., Rimm, E.B., Morris, J.S., et al., 2002. N. Engl. J. Med.
miol. 163, 694–699. 347, 1755–1760.
Taneja, S.K., Mandal, R., 2007. Biol. Trace Elem. Res. 120, 61–73. Zhang, W., Iso, H., Ohira, T., the JACC study group, 2012a. J. Epide-
Tanguy, S., Grauzam, S., de Leiris, J., et al., 2012. Mol. Nutr. Food Res. miol. 22, 484–493.
56, 1106–1121. Zhang, W., Iso, H., Ohira, T., et al., 2012b. Atherosclerosis 221,
Tellez-Plaza, M., Navas-Acien, A., Crainiceanu, C.M., et al., 2008. 587–595.
Environ. Health Perspect. 116, 51–56. Zimmermann, M.B., Hurrell, R.F., 2007. Lancet 370, 511–520.
Tellez-Plaza, M., Navas-Acien, A., Menke, A., et al., 2012. Environ. Zivin, J.A., 2004. Approaches to cerebrovascular diseases. In:
Health Perspect. 120, 1017–1022. Goldman, L., Ausiello, D. (Eds.), Cecil Textbook of Medi-
Tellez-Plaza, M., Guallar, E., Howard, B.V., et al., 2013a. Epidemiol. cine, twenty second ed. Saunders, Philadelphia, PA, USA,
24, 421–429. pp. 2280–2298.

You might also like