You are on page 1of 12

REVIEWS

Environmental pollution and kidney


diseases
Xin Xu, Sheng Nie, Hanying Ding and Fan Fan Hou*
Abstract | The burden of disease and death attributable to environmental pollution is becoming a
public health challenge worldwide, especially in developing countries. The kidney is vulnerable to
environmental pollutants because most environmental toxins are concentrated by the kidney
during filtration. Given the high mortality and morbidity of kidney disease, environmental risk
factors and their effect on kidney disease need to be identified. In this Review, we highlight
epidemiological evidence for the association between kidney disease and environmental
pollutants, including air pollution, heavy metal pollution and other environmental risk factors.
We discuss the potential biological mechanisms that link exposure to environmental pollutants
to kidney damage and emphasize the contribution of environmental pollution to kidney disease.
Regulatory efforts should be made to control environmental pollution and limit individual
exposure to preventable or avoidable environmental risk. Population studies with accurate
quantification of environmental exposure in polluted regions, particularly in developing
countries, might aid our understanding of the dose–response relationship between pollutants
and kidney diseases.

Kidney disease has been increasingly recognized as a environment. The kidney is especially vulnerable to
public health problem owing to its increasing prevalence, environmental pollutants, as almost 20% of the cardiac
the increased risk of cardiovascular disease (CVD) and output of blood is delivered to the kidneys, where the
death in patients with kidney disease and the high cost blood is filtered and environmental toxins can be con-
of treatment. A meta-analysis of 33 studies estimated centrated. Although diabetes, hypertension and primary
that the worldwide prevalence of chronic kidney disease glomerulonephritis are major causes of CKD in most
(CKD) in 2010 was 10.4% in men and 11.8% in women1, middle-income and high-income countries, infectious
with the majority of patients being from developing diseases and environmental and occupational exposure
countries. The Global Burden of Disease Study 2015 to pollutants remain common causes of kidney disease
estimates that 1,234,900 deaths in 2010 were directly in the developing world.
attributable to CKD, which represents a 31.7% increase In this Review, we summarize the epidemiological
from 2005 (REF. 2). The increasing prevalence of CKD evidence for the association between kidney disease
and the variation in the burden of kidney disease cannot and exposure to environmental pollutants, including
be fully explained by trends in traditional drivers, such air pollution, heavy metal pollution and other potential
as diabetes mellitus and hypertension, suggesting that environmental risk factors. We also discuss the poten-
other previously unappreciated risk factors contribute tial biological mechanisms that might link exposure to
National Clinical Research
to the disease process3–6. ­environmental toxins to kidney damage.
Center for Kidney Disease, The effects of increasing environmental pollution
State Key Laboratory of (of air, water and soil, typically by chemical products Air pollution and kidney disease
Organ Failure Research, in daily use), which is a result of accelerated industri- The Global Burden of Disease Study estimates that
Nanfang Hospital, Southern
alization and urbanization worldwide, have become a 6.4 million deaths in 2015 were attributable to air pol-
Medical University, 1838
North Guangzhou Avenue, global health challenge. The World Health Organization lution2. Epidemiological and experimental studies have
Guangzhou 510515, China. (WHO) reported that preventable environmental risks clearly established that air pollution, and particulate mat-
*e-mail: accounted for 12.6 million deaths worldwide and 22% ter from both pulmonary and extrapulmonary sources in
ffhouguangzhou@163.com of the global burden of disease in 2012 (REF. 7) (FIG. 1). particular, contributes to cardiovascular morbidity and
doi:10.1038/nrneph.2018.11 More than 1.7 million children younger than 5 years mortality 8. Particulate matter is generally categorized
Published online 26 Feb 2018 of age die every year as a result of living in a polluted by its mean aerodynamic diameter as PM10 (particulate

NATURE REVIEWS | NEPHROLOGY VOLUME 14 | MAY 2018 | 313


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points In humans, most inhaled particles >5 μm in aero-


dynamic diameter are trapped in the fluid that lines
• Up to 22% of the global burden of disease and 23% of deaths are attributable to the upper airways or larger lower airways, are moved
environmental pollution; the general public is inevitably exposed to environmental upwards by the mucociliary ladder and are excreted from
pollutants the body or swallowed. Particles <5 μm in aerodynamic
• The kidney is particularly vulnerable to toxic effects from environmental pollutants diameter are more likely to be deposited in the bron-
owing to its filtration functions; environmental and occupational exposures to chioles and the alveoli of the lung 20. Particles deposited
pollutants remain common causes of kidney disease worldwide, especially in
beyond the terminal bronchioles are cleared by lung mac-
developing countries
rophages that, in turn, transport the ingested particles
• Long-term exposure to particulate matter <2.5 μm in mean aerodynamic diameter
onto the mucociliary ladder or into the lymphatic sys-
(PM2.5) is associated with increased risk of membranous nephropathy and more rapid
decline in renal function
tem. Alveolar phagocytes and pulmonary dendritic cells
engulf any extraneous materials deposited at the lung
• Exposure to heavy metals leads to acute and chronic kidney injury; tubular
dysfunction is the most common manifestation of nephrotoxicity from heavy metals
epithelium, efficiently but slowly removing the foreign
particulate matter 21–23.
• Owing to the worldwide distribution of Aristolochia spp. and the still widespread use
of medicinal herbal remedies containing aristolochic acids, especially in east Asia, it is
Adverse effects of particulate matter on the lungs
possible that aristolochic acids might be the cause of unrecognized nephropathies include disruption of the airway epithelial barrier and
cellular signalling pathways24–26, induction of oxidative
• Exposure to industrial and agricultural chemicals, biogenic toxins and secondhand
smoke are additional risk factors for kidney disease stress27–30, impairment of phagocytosis28, infiltration of
inflammatory cells, and dysregulation of cellular immu-
nity, epigenetic modifications and autophagy 27,28. The
matter <10 μm in diameter), PM2.5 (<2.5 μm) or ultra­ pulmonary inflammatory mediators induced by PM2.5
fine particles (UFPs, <0.1 μm) (FIG. 2). Exposure to PM2.5 could spill over into the circulation, resulting in systemic
caused 4.2 million deaths in 2015, which represents inflammation, oxidative stress and damage to distant
7.6% of deaths worldwide, and contributed to 17.1% of organs8. PM2.5 deposited in alveoli activates an autonomic
mortality from ischaemic heart disease, 14.2% of mortal- nervous system reflex, which induces an autonomic nerv-
ity from cerebrovascular disease and 16.5% of mortality ous system imbalance31 and promotes systemic oxidative
from lung cancer. Approximately 60% of PM2.5-related stress and other organ damage. Furthermore, deposited
deaths occurred in south and east Asia9,10. ultrafine particles may translocate directly into the circu-
Air pollution is a complex mixture of gaseous com- lation or the lymphatic system, leading to dysfunction of
ponents and solid and liquid particles suspended in the coagulation and/or fibrinolysis and, subsequently, cellu-
air and can vary substantially in chemical composition lar responses in nonpulmonary tissues32, which explains
between different cities. Particulate matter, which pri- why people with pre-existing disease are particularly
marily comprises solid particulates that are derived vulnerable when exposed to particulate matter 33,34 (FIG. 3).
from the combustion of coal, gasoline and diesel fuels, Of note, inhaled particles <30 nm in diameter can trans­
is the major element of air pollution that causes the locate from the lungs into the circulation, are filtered and
most adverse health effects in animals and humans8,11,12. excreted by the kidneys and selectively accumulate at sites
In large cities, the majority of particulate matter pro- of vascular inflammation in both animals and healthy
duction comes from road traffic and industrial burning men35. Whether the deposition of inhaled nanoparticles
of fossil fuels13. contributes to the progression of cardiovascular disease
Gaseous compounds, such as nitrogen dioxide, car- in patients with CKD requires further investigation.
bon monoxide, ozone and sulfur oxides, are another Mounting evidence implicates air pollution in the
important component of air pollution. Nitric oxides and pathogenesis of pulmonary and cardiovascular diseases,
carbon monoxide are mainly derived from road traffic including asthma, myocardial infarction, stroke, cardiac
and the industrial burning of fuels14, whereas the indus- arrhythmia and heart failure. A comprehensive review
trial production of sulfur-based products is the major of the likely biological mechanisms strongly supports
source of sulfur dioxide15. Exposure to these gaseous a causal relationship between air pollution and cardio­
pollutants is widely acknowledged to cause various vascular disease and mortality 4. Although currently limi­
health problems in adults, including chronic obstruc- ted, data on the link between air pollution and kidney
tive pulmonary disease, asthma, cataracts and blind- injury or disease in humans and in animal models are
ness, whereas gaseous pollutants cause acute respiratory also now beginning to emerge.
­infections in both adults and children16.
Air quality varies markedly between different coun- Epidemiological studies. A study of 1,103 consecutive
tries, different cities within a country and even different patients who were hospitalized with acute ischaemic
locales within a city. In the decade from 2000 to 2010, stroke in the Boston (MA, USA) metropolitan region
urban expansion in east Asia has exacerbated air pollu- found an association between the proximity of a patient’s
tion17. For example, the 1‑year average of PM2.5 exceeds home to a major roadway and their estimated glomer-
50 μg/m3 in developing countries such as China and India ular filtration rate (eGFR)36. Individuals living 50 m
compared with 10–14 μg/m3 in more-developed coun- from a major roadway had a 3.9 ml/min/1.73 m2 lower
tries such as the USA, the UK and Japan18. The PM2.5 val- eGFR than those who lived 1,000 m away. In another
ues ranged from 6 μg/m3 to 114 μg/m3 among 282 cities cohort study that included 669 elderly men from the
in China, with an interquartile range of 41 μg/m3 (REF. 19). Boston area, 1‑year PM2.5 exposure was associated with

314 | MAY 2018 | VOLUME 14 www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Disease burden (%)


9–14
15–16
17–18
19–22
23–31
Data not available
Not applicable
0 850 1,700 3,400
Kilometres

Figure 1 | Worldwide distribution of disease burden attributable to environmental risks in 2012. As a proportion of
Nature Reviews
total disease burden, the burden of disease attributable to environmental risks is higher in low-income | Nephrology
and middle-income
countries than in high-income countries. Reprinted from World Health Organization, Prüss-Ustün, A., Wolf, J., Corvalán, C.,
Bos, R. & Neira, M. Preventing disease through healthy environments: a global assessment of the burden of disease from
environmental risks, copyright (2016), REF. 7.

a lower eGFR and a faster annual decline of eGFR37. without registry data for the biopsy samples, the aggre-
Each 2.1 μg/m3 increase in PM2.5 exposure was associ- gated effect of PM2.5 on kidney disease in the general pop-
ated with a 1.87 ml/min/1.73 m2 reduction in eGFR and a ulation cannot be estimated. Furthermore, the study
0.60 ml/min/1.73 m2 per year additional decline in annual population is characterized by exposure to unusually high
renal function. levels of PM2.5, so the results might not be generalizable to
A nationwide study of renal biopsy samples obtained a population exposed to low levels of PM2.5.
during 2004–2014 included 71,151 native kidney biopsy A large cohort study of >2 million US veterans with no
samples from 938 hospitals in 282 cities across China19. previous history of kidney disease found that long-term
During the study period, the level of PM2.5 exposure var- exposure to PM2.5, PM10, nitrogen dioxide and carbon
ied from 6 μg/m3 to 114 μg/m3 (mean 52.6 μg/m3) among monoxide is associated with an increased risk of incident
the 282 cities. The researchers found that the prevalence CKD, CKD progression and development of end-stage
of membranous nephropathy increased from 12% to 24% renal disease (ESRD)38. Each 10 μg/m3 increase in PM2.5
during the decade, whereas the prevalence of other major concentration was associated with 26–28% higher risk of
glomerular diseases remained relatively stable. More strik- incident CKD, CKD progression and ESRD. The effect
ingly, long-term exposure to high levels of PM2.5 was asso- sizes of different types of air pollutants (PM2.5, PM10,
ciated with an increased risk of membranous nephropathy nitrogen dioxide and carbon monoxide) were generally
in a nonlinear pattern. Each 10 μg/m3 increase in PM2.5 comparable, and the relationship between the exposures
concentration was associated with 14% higher odds of a and the risk of CKD seemed to be linear in the range
patient developing membranous nephropathy (OR 1.14, studied. The researchers estimated that approximately
95% CI 1.10–1.18) in regions with PM2.5 levels >70 μg/m3. 10–13% of the cases of CKD in the USA might be attrib-
The effect size in regions with PM2.5 <60 μg/m3 was greatly utable to air pollution. Of note, the air quality in the
attenuated. The findings from this study suggest that USA is relatively good compared to that of other coun-
patients with long-term exposure to PM2.5 are more tries, with a median exposure to PM2.5 of 10–11 μg/m3.
likely to develop membranous nephropathy than they Establishing the dose–response relationship between
are to develop other kidney diseases. However, the study air pollution and the development and progression of
included only patients from whom renal biopsy samples CKD across a wide range of exposure levels is of great
were taken and not the general population. Therefore, clinical importance.

NATURE REVIEWS | NEPHROLOGY VOLUME 14 | MAY 2018 | 315


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

inflammatory signalling and endothelial dysfunction,


in the kidneys49. These results provide insights into the
potential mechanisms that underlie the health effects of
ozone inhalation and the susceptibility of the kidneys.
The risk of developing membranous nephropathy
has been associated with the level of exposure to PM2.5
(REF. 19), but the mechanisms underlying this association
remain to be elucidated. Membranous nephropathy is an
autoimmune disease that is characterized by the forma-
0 50 100 tion of circulating autoantibodies against secretory phos-
Particle size (μm)
pholipase A2 receptor (PLA2R; encoded by PLA2R1) and
PM2.5 (combustion particles, PM50–70 (human hair) deposition of immune complexes at the glomerular base-
organic compounds, metals, etc.) ment membrane50. M‑type PLA2R, which is expressed by
PM90 (fine beach sand)
PM10 (dust, pollen, mould, etc.) alveolar macrophages51 and glomerular podocytes52, has
been recognized as the major antigen that is implicated
Figure 2 | Size classification of particulate matter. Particulate matter
Nature (PM) in
Reviews airborne
| Nephrology in idiopathic membranous nephro­pathy, accounting for
pollutants is classified based on the mean aerodynamic diameter of particles into PM10 ~70% of cases of idiopathic membranous nephropathy 52.
(<10 μm), PM2.5 (<2.5 μm) and ultrafine particles (<0.1 μm). The sizes of various biological
Exposure to particulate matter might alter the structure
entities are depicted for comparison.
of PLA2R in the lung and stimulate the generation of
autoantibodies against PLA2R. The circulating auto­
Mechanical studies. The pathogenic mechanisms under- antibodies interact with PLA2R on glomerular podo-
lying the association between air pollution and kidney dis- cytes, inducing the formation of immune complexes
ease remain to be completely elucidated. Diesel exhaust that damage the cells, which then results in membra-
particles (DEPs) are the major source of PM2.5 and UFPs nous nephropathy. Animal studies support this mecha-
in densely populated urban regions, contributing an esti- nism of particulate matter toxicity — exposure of mice
mated 30% of the total particulate matter in the atmos- to fine particulate matter promotes formation of auto­
phere39,40. In rodent models of acute kidney injury induced antibodies and immune complexes53. Interestingly, cir-
by cisplatin, both single and repeated exposure to DEPs culating autoantibodies against PLA2R were detect­able
in the lung aggravated cisplatin-induced nephrotoxicity, in approximately 80% of patients with membranous
resulting in increased urinary excretion of protein and nephropathy in a cohort study 19. It is important to exam-
N‑acetyl-β‑d‑glucosaminidase, impaired renal func- ine whether the association between particulate matter
tion and increased severity of renal tubular necrosis41,42. and ­membranous nephropathy is antigen-specific.
In rodents with adenine-induced CKD, exposure to DEPs A study in traffic policemen demonstrated that expo-
resulted in a substantial decrease in renal blood flow and sure to PM2.5 increased the circulating levels of inflam-
an increase in renal oxidative stress, inflammation matory mediators such as tumour necrosis factor (TNF)
and DNA damage43–46. The nephrotoxicity of DEPs could and IL‑6 (REF. 54). An explanation for this effect is that
be partially abrogated by administration of the antioxidant cytokines generated in the airways in response to PM2.5
thymoquinone46. A pathogenic effect of DEPs in human might spill over into the circulation, inducing systemic
diseases, including diseases of the respiratory system and inflammation and oxidative stress, and thus promote
distant organs such as the heart and the kidneys, has been distant organ damage.
reported47, although the mechanism remains unclear. Genetic factors have been implicated in suscepti­
Further study of the composition of different sources of bility to idiopathic membranous nephropathy. Several
air pollution is required to improve our understanding risk alleles, such as the PLA2R1, HLADQA1 and
of the adverse health effects of air pollution on the kidney. HLADRB1 loci, are strongly associated with an increased
Ozone is a product of the interaction between ultra­ risk of PLA2R‑related membranous nephropathy 55–57.
violet light and oxides of nitrogen and volatile hydro- Additional studies are needed to determine the relation-
carbons. Short-term inhalation of ozone provokes an ship between genetic background and environment pol-
inflammatory and metabolic response in the lungs lutants and the role of genetic factors in the pathogenesis
and also in extrapulmonary organs, such as the kid- of idiopathic membranous nephropathy.
neys. Inhalation of ozone activates the hypothalamic–­
pituitary–adrenal axis in rats and increases plasma levels Heavy metals and kidney disease
of the glucocorticoid corticosterone. Several immuno­ Human activities, such as mining, industry, urban
suppressive and metabolic effects of ozone in the lungs, expansion and the use of fertilizers in agriculture, release
heart, liver, kidneys and spleen are blocked by metyrapone substantial quantities of pollutants into the environment
(an inhibitor of cortisol production) and are reproduced and ecosystems. Among all types of pollution in drink-
through exogenous administration of corticosterone, ing water and food, heavy metals are considered the
demonstrating glucocorticoid-dependent effects in tar- greatest threat to human health owing to their persis-
get tissues48. In addition, gene expression analyses in rats tence in the environment and their bioaccessibility, as
showed that inhalation of ozone (0.4 ppm and 0.8 ppm) they can be absorbed into the human body from the diet
resulted in altered expression of genes that are involved and drinking water, by inhalation and even by dermal
in various pathways, including the antioxidant response, contact58,59.

316 | MAY 2018 | VOLUME 14 www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

A large proportion of the world’s water is polluted first occurs in plants, then in animals and eventu-
with heavy metals, which in turn contaminates the soil ally in humans, leading to disorders and diseases60,61.
through irrigation, followed by contamination of the Approximately 10% of farmland soil and 13.86% of
crops planted in these soils and subsequently the animals grain production in China is estimated to be contami-
and humans further up the food chain. Accumulation nated with heavy metals through irrigation and the use

PM2.5 inhalation

Lung
• ↑ Oxidative stress and
inflammation
• Impairment of phagocytosis
• Dysregulated cell immunity
• Epigenetic modification
• Disruption of cellular
signalling pathways
Nervous system
PM2.5 • ANS imbalance
Alveolus • ↑ Sympathetic nervous system
Macrophage • ↓ Parasympathetic nervous system
Cytokine
release PM uptake Activation of
ANS reflex

Systemic Circulation PM transmitted


spillover into circulation

Systemic inflammation and oxidative


stress Coagulation and fibrinolysis
• ↑ Cellular response (activated dysfunction
leukocytes and platelets and increased • Altered rheology
MPO) • ↑ Coagulability
• ↑ Cytokine expression (IL-6, IL-8, IL-1β, • ↑ Peripheral thrombosis
TNF) • ↓ Oxygen saturation
• ↑ ROS-generating pathway (activation • ↓ Fibrinolysis
of NADPH oxidases) • ↑ Platelet aggregation
• ↓ Antioxidants

Kidney
• Inflammation and oxidative stress
• Circulating autoantibody
against PLA2R
• Formation of immune complexes
• Vascular (endothelial) injury
Vasculature
• Vasoconstriction
• Endothelial dysfunction
Heart • ↑ ROS
• ↑ Susceptibility to dysrhythmia
• Altered cardiac autonomic function
• Altered cardiac repolarization

Figure 3 | Potential mechanisms linking inhaled particles to pulmonary, cardiovascular, cerebral and kidney diseases.
Deposition of particulate matter (PM) with a mean aerodynamic diameter <2.5 μm (PM2.5) in the bronchioles and alveoli
Nature Reviews
leads to disruption of the airway epithelial barrier and to altered cellular signalling pathways, oxidative | Nephrology
stress and
inflammation. The pulmonary inflammatory mediators induced by PM2.5 might spill over into the circulation, resulting
in systemic inflammation, oxidative stress and damage to other distant organs. PM2.5 deposited in alveoli activates an
autonomic nervous system (ANS) reflex, inducing ANS imbalance and promoting damage to other organs. Furthermore,
ultrafine particles (<0.1 μm in diameter) might translocate directly into the circulation, leading to coagulation and/or
fibrinolysis dysfunction and, subsequently, to cellular responses in nonpulmonary tissues. MPO, myeloperoxidase;
PLA2R, secretory phospholipase A2 receptor; ROS, reactive oxygen species; TNF, tumour necrosis factor.

NATURE REVIEWS | NEPHROLOGY VOLUME 14 | MAY 2018 | 317


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

of fertilizers62,63. Exposure to these pollutants via drink- dietary sources, including mining, fuel combustion,
ing water represents a major health risk, especially for drinking contaminated water, eating freshwater fish
people living in developing countries because of a lack species from polluted waters, eating predatory ocean
of sufficient water treatment facilities64–66. It is estimated fish species and the use of skin-whitening creams88.
that >1.8 billion people in the world do not have access Although the major toxicity associated with mercury
to safe drinking water 67,68. exposure is neurological and digestive, nephrotoxicity
has also been reported89. As inorganic mercury is pre-
Cadmium. Cadmium is a by-product of mining and dominantly reabsorbed in the proximal tubules, they
is used industrially for the plating of steel and in the are the sites that are most sensitive to mercury nephro­
production of plastics and nickel–cadmium batteries. toxicity. Acute effects of mercury, including mitochon-
Repeated use of agrochemicals in agriculture, especially drial injury and DNA damage, have been observed in
phosphate fertilizers, causes deposition of cadmium human proximal tubular cells90,91. Acute exposure to
in intensively farmed soils, eventually resulting in its mercury can cause tubular epithelial cell necrosis and
transportation to and accumulation in the plant shoot 69. low-molecular-weight proteinuria92.
In developing countries such as China, cadmium is one Of note, chronic exposure to mercury also has detri­
of the major heavy metal pollutants in farmland70. Diet mental effects on glomeruli. In a systematic review of
is the primary source of cadmium exposure, followed by 27 case reports of 42 patients with mercury-associated
tobacco smoke71,72. Cadmium has a long biological half- nephrotic syndrome, 26 patients underwent a renal
life in the human body of 7–16 years73 and accumulates biopsy 93. Among these 26 patients, 21 (80.7%) had
in various organs and tissues, particularly in the renal glomerular diseases, which included membranous
cortex 74. In Asian countries, rice is a staple food and cad- nephropathy (15/26), minimal change disease (4/26),
mium accumulation in rice has been identified as the focal segmental glomerulosclerosis (1/26) and chronic
leading source of cadmium burden, which is associated proliferative glomerulonephritis (1/26). Another case
with renal dysfunction in these areas75. series from China enrolled 11 patients with mercury-­
Studies of populations that are exposed to low levels induced membranous nephropathy 94. The duration
of cadmium showed that cadmium is present in children of exposure to mercury in these patients ranged from
and even in infants and that the cadmium concentra- 2 months to 5 years, and the urinary mercury concen-
tion increases in adults and reaches its peak in elderly trations were 1.5–50 times higher than in individuals
individuals76. Urinary cadmium levels have been used as without exposure to mercury. Unlike in patients with
a biomarker of the extent of ongoing and chronic cad- idiopathic membranous nephropathy, patients
mium exposure in the general population. Urinary with  mercury-­induced membranous nephropathy
cadmium levels of 4–10 μg/g creatinine are associated were nega­tive for the PLA2R‑specific autoantibody in
with increased microalbuminuria77. Accordingly, the serum95. In addition, immunoglobulin G1 (IgG1) is the
US Occupational Safety and Health Administration set predominant immunoglobulin in glomerular deposits
a urinary cadmium concentration of <3 μg/g creatinine in patients with mercury-induced membranous nephro­
as the safety standard, whereas the WHO set the safe pathy, whereas IgG4 is the primary glomerular deposit
threshold of urinary cadmium at 5.24 μg/g creatinine78. in patients with idiopathic membranous nephropathy 94,
The best-known case of cadmium poisoning is itai- suggesting that the pathogenesis differs between the
itai disease, which was caused by contamination of the two forms of membranous nephropathy. Interestingly,
Jinzu River in Japan due to mining activity 79. The river >80% of the patients achieved complete remission after
water was used for irrigation of rice fields, the rice plants ­withdrawal from m ­ ercury exposure.
efficiently absorbed the cadmium, and ingestion of the The pathogenic mechanisms of mercury-induced
contaminated rice resulted in the accumulation of toxic glomerular disease have not been fully elucidated. In
levels of cadmium in people. The affected individuals animal models, mercury induces autoimmune dys-
suffered from anaemia, severe bone pain, osteomalacia function that is characterized by activation of T cell-­
and kidney failure. dependent polyclonal B cells, increased serum levels of
The proximal tubule and glomeruli are the major IgG and IgE, production of antinuclear autoantibodies
targets in cadmium-induced nephropathy, as cadmium (ANAs) and formation of immune complex deposits in
is preferentially taken up by the proximal tubules and the kidneys96–100. A higher prevalence of detectable ANAs
mesangial cells in glomeruli80–82. The nephrotoxicity of and increased serum concentration of inflammatory
cadmium is dependent on the dose: one-time exposure cytokines, such as IL‑1β, TNF and IFNγ, are observed
to cadmium induces reversible low-molecular-weight in gold miners with chronic exposure to mercury 101.
proteinuria, whereas chronic repeated exposure causes Several autoantibodies have been identified as novel
irreversible proteinuria83,84. Chronic cadmium exposure potential biomarkers of mercury-induced immuno­
results in kidney injury through several mechanisms, toxicity, including those that react with glutathione
including oxidative stress85, inhibition of the repair of S‑transferase α1 (GSTA1), TNF ligand superfamily mem-
oxidative DNA damage86 and apoptosis87. ber 13 (TNFSF13) and linker for activation of T cells fam-
ily member 1 (LAT)102. Mercury-induced auto­immunity
Mercury. Mercury is a widespread heavy metal pol- and inflammation might result in the formation of
lutant. Humans are exposed to mercuric compounds circulating immune complexes that deposit in the kid-
predominantly from occupational, environmental and neys, or it might lead to the production of antibodies

318 | MAY 2018 | VOLUME 14 www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

against membrane proteins on podocytes. Testing of Earth’s crust, with an average concentration of 75 μg/g
these hypotheses of mercury-induced immunotoxicity (REF. 127). Compounds containing nickel are widely used
is crucial for understanding the role of ­mercury in the in industrial and commercial areas, and extensive indus-
pathogenesis of autoimmune renal diseases. trialization has resulted in the release of excess nickel into
ecosystems. Nickel has genotoxic, immunotoxic, muta-
Lead. Lead is a chemically stable heavy metal with a genic and carcinogenic effects in humans128. The kidney
low melting point and poor electrical conductivity, and is the major organ for nickel accumulation and excretion.
thus it is widely used in industrial products. Exposure to Excess nickel triggers an inflammatory response via acti-
lead primarily occurs through diet and drinking water, vation of nuclear factor‑κB (NF‑κB) and tubular apopto-
and to a lesser extent by inhalation, and occasionally by sis through the phosphoinositide 3‑kinase (PI3K)–RAC
dermal contact with organic lead compounds. In adults, serine/threonine-protein kinase (AKT) pathway 129,130.
most of the lead that enters the body is excreted in the Sources of heavy metal pollutants, the clinical features
urine, whereas children excrete only 32% of internalized of pollution-related kidney diseases and possible patho­
lead. Lead is one of the most toxic heavy metals, as it can genic mechanisms have been reviewed elsewhere131–143
remain in the soil for over 150 years. A portion of the lead (summarized in TABLE 1).
that enters the body cannot be excreted in the urine and
is deposited in bone, where its half-life is 10–30 years. Other environmental pollutants
Of note, lead can cross the blood–placenta barrier freely, Biogenic toxins. Aristolochic acids are a family of
and thus maternal lead exposure can affect a fetus103. carcinogenic and nephrotoxic compounds that are
Exposure to even a low dose of lead in infancy and commonly present in members of the birthwort
early childhood can result in impairment of c­ ognitive, (Aristolochiaceae) family of plants. The family includes
­neurochemical and behavioural development104–106. the Aristolochia and Asarum (wild ginger) genera.
Data from population-based studies showed that Aristolochic acids are the causative agents of Balkan
chronic exposure to lead increases the incidence of CKD endemic nephropathy, which is a chronic, progressive
and nephrolithiasis107–109. Lead exposure causes injury tubulointerstitial disease that is clustered in the residents
in proximal tubular cells, glomerular dysfunction and of rural farming villages located along tributaries of the
interstitial fibrosis, with clinical manifestations such as Danube River in southeastern Europe144. An unusual
albuminuria and reduced GFR110–113. Lead-induced mito- feature of Balkan endemic nephropathy is the strikingly
chondrial swelling and energy metabolism dysfunction high incidence (>50%) of upper urinary tract cancers
in tubular cells also contribute to the pathogenesis of in patients145. Balkan endemic nephropathy was first
kidney disease114. The accumulation of lead in the kid- described in the 1950s and exhibits a familial but not
ney results in upregulated transforming growth factor-β inherited association, suggesting the importance of envi-
(TGFβ) expression and lipid peroxidation, leading to the ronmental factors. Members of the Aristolochiaceae fam-
development of tubulointerstitial lesions and fibrosis115,116. ily grow abundantly as weeds in local wheat fields around
the towns affected by Balkan endemic nephro­pathy, and
Other heavy metals. Arsenic is an element that occurs their seeds are co‑mingled with wheat grain during the
naturally in soil, food and water. Humans are exposed to harvesting process. Balkan endemic nephro­pathy is
inorganic arsenic from mining and smelting metal ores, now believed to be caused primarily by consumption of
pesticide manufacture, wood preservatives and medi­ aristolochic acids present in flour obtained from wheat
cines. Food may contain both organic and i­norganic grown in fields contaminated with Aristolochia clema-
arsenic, whereas drinking water contains primarily titis146. Kidney failure also occurred among hundreds
inorganic arsenic 117–119. People living in areas with of Belgian women who received a Chinese herbal rem-
major arsenic contamination usually have an increased edy containing aristolochic acids, which confirmed the
incidence of arsenic-related cancers, such as carcino- nephrotoxicity of these compounds 147. Owing to
mas of the liver, lung, skin, bladder and kidney 120,121. the worldwide distribution of Aristolochia spp. and the
Studies of arsenic-induced nephrotoxicity are rare, but still widespread use of medicinal herbal remedies con-
one study reported an increased risk of kidney disease in taining aristolochic acids, especially in east Asia, it is
individuals exposed to arsenic-contaminated drinking possible that aristolochic acids might be the cause of
water in MI, USA122. ­nephropathies of unrecognized aetiology148.
Although copper is an essential trace element, The pathogenic mechanisms of aristolochic acid-­
excessive intake of copper can be toxic owing to its induced nephropathy are not fully understood. In both
redox-­active nature, which induces oxidative stress acute and chronic aristolochic acid-induced nephro­
and thereby cellular injury in organs in which copper is pathy, autophagy can be activated via upregulation of
deposited123. Copper in water and soil enters the human the extracellular signal-regulated kinase 1 (ERK1; also
body through food intake and reaches the kidneys via known as MAPK3) and ERK2 (also known as MAPK1)
the circulation124. Copper nephrotoxicity is characterized pathways149 and by induction of autophagy protein 5
by proximal tubule necrosis that results from oxidative (ATG5) and microtubule-associated proteins 1A/1B
stress, which is induced by copper catalysing the forma- light chain 3B (MAPILC3B; also known as LC3) expres-
tion of highly reactive hydroxyl radicals125,126. Nickel is sion 150. In addition, administration of aristolochic
an essential element, as it is a constituent of enzymes, acid triggers the upregulation of proteins involved in
proteins and nucleic acids. Nickel is abundant in the mitochondrial and endoplasmic reticulum stress149–151.

NATURE REVIEWS | NEPHROLOGY VOLUME 14 | MAY 2018 | 319


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Aristolactam, a metabolite of aristolochic acid, forms is a widely used herbicide that induces glomerular
DNA adducts in  the kidney and causes mutations lesions and renal tubular necrosis in animal studies165.
in TP53, which encodes the tumour suppressor p53 In a large prospective study of individuals involved in
(REF. 152). Furthermore, the TP53 mutations caused by applying pesticides in the USA166, exposure to six pesti­
aristolochic acid are dominated by A>T to T>A trans- cides, including five herbicides (paraquat, alachlor,
versions, and the TP53 gene contains several unique hot atrazine, metolachlor and pendimethalin) and an insec-
spots (‘mutation signatures’) that are highly specific for ticide (permethrin), was associated with increased risk
aristolochic acid-induced carcinogenicity 153–156. of ESRD. Intriguingly, risk of ESRD was also increased
Ochratoxin A (OTA) is one of the most common among the spouses of those applying the paraquat, who
mycotoxins and is a ubiquitous natural contaminant of do not come in direct contact with the pesticides167.
food, including wheat, nuts, spices, dried meats, milk, Chronic exposure to an ultra-low dose of glyphosate in
fruit juices, wine and cocoa. Removing or reducing an established animal toxicity model system can result
the level of OTA in food processing is difficult. Animal in kidney damage168. In a case–control study of patients
studies from the US National Cancer Institute–National with CKD, deficiencies in the activity of the glutathione
Toxicological Program (NCI–NTP) have established that S‑transferases GSTθ1 and GSTμ1 were associ­ated
OTA is nephrotoxic and has renal carcinogenic effects157. with accumulation of organochlorine pesticides and
Exposure to OTA can lead to renal disorders, such as ­aggravation of kidney dysfunction169.
acute kidney injury and upper urothelial carcinoma. Only a limited number of the thousands of pesticides
OTA is also considered to be a potential cause of Balkan used worldwide have been tested for nephrotoxicity. The
endemic nephropathy in the Balkan peninsula and of heterogeneity in study design and unquantified assess-
chronic interstitial nephropathy in northern African ment of pesticide exposure limit the interpretation of
countries158,159. The mechanisms of OTA nephrotoxicity results from published studies. Additional studies are
include inhibition of protein synthesis, lipoperoxidation needed to characterize the nephrotoxicity of pesticides
and modulation of the mitogen activated protein kinase and their pathogenic mechanisms.
(MAPK) cascade160.
Industrial chemicals. The general public is unknow-
Agricultural pesticides. Pesticides are chemicals that are ingly exposed to industrial chemicals, such as phtha-
used to destroy destructive pests or to prevent or control lates, bisphenol A and perfluoroalkyl acids, during daily
their growth. Approximately 3 million tons of pesticides, consumer activities. These chemicals are widely used
mainly herbicides, insecticides and fungicides, are used in shampoo, cosmetics and food packaging. Numerous
globally every year. Exposure to pesticides might cause cross-sectional studies have linked exposure to these
various health problems, ranging from irritation of the industrial chemicals with an increased incidence of
skin and eyes to more severe effects, such as nervous kidney disease170. The melamine poisoning incident in
system disorders, reproductive problems and cancer 161. 2009 exemplifies the devastating effect of widespread
A number of widely used pesticides are established exposure to chemical contaminants on human health
human nephrotoxins, including paraquat 162, glypho- and, in particular, kidney disease. Exposure of >300,000
sate163 and some organochlorine insecticides164. Paraquat infants and children in China to milk-based formulas

Table 1 | Effects of heavy metal pollution on the kidney


Metal Source of exposure Kidney injury Mechanisms
Cadmium Contaminated food (rice); Proximal tubular dysfunction Oxidative stress85; impaired
cigarette smoke; industrial waste; (glucosuria, aminoaciduria DNA repair86; reduced
occupational exposure (mining, and low-molecular-weight antioxidant ability; cellular
production of batteries, plating of proteinuria)83,84,131; reduced GFR apoptosis87
steel and plastic manufacturing)
Lead Contaminated food; petroleum; Proximal tubular Oxidative stress134; increased
contaminated air, water and soil dysfunction110–113; interstitial TGFβ expression and lipid
polluted with industrial waste; fibrosis132; tubular atrophy133; oxidation115,116; mitochondrial
cigarette smoke; occupational reduced GFR dysfunction114; DNA
exposure (mining, production of fragmentation135
batteries, welding and lead soldering)
Mercury Contaminated water; fish from Secondary membranous DNA damage90,91; mitochondrial
polluted waters; fuel combustion; nephropathy136; interstitial dysfunction138; reduced
skin-whitening creams; mining nephritis; acute tubular enzymatic activity139
necrosis137; reduced GFR
Arsenic Occupational exposure (mining, Tubular interstitial nephritis; Oxidative stress; reduced
wood preservatives, smelting metal acute tubular necrosis140; expression of RKIP141; DNA
ores and pesticides); contaminated reduced GFR methylation and histone
seafood and water; specific acetylation142; DNA oxidation;
medication reduced antioxidant defences143
GFR, glomerular filtration rate; RKIP, RAF kinase inhibitor protein; TGFβ, transforming growth factor‑β.

320 | MAY 2018 | VOLUME 14 www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

containing melamine resulted in kidney stone forma- Conclusions


tion, renal dysfunction and retarded renal growth in Environmental pollutants, such as particulate matter,
these children171,172. heavy metals and industrial and agricultural chemicals,
are important risk factors for kidney disease, especially
Secondhand smoke in children. Tobacco smoke contains in developing countries in which environmental pollu-
>4,000 toxic chemicals and particles that are nephrotoxic, tion is prevalent. The causes of CKDu and the patho-
including nicotine, carbon monoxide, arsenic, cadmium genic mechanisms of most environmental nephrotoxins
and lead. Cigarette smoking is associated with increased remain to be elucidated. As mentioned earlier, approxi-
risk of CKD and ESRD in adults. A meta-­analysis of mately 10–13% of CKD cases in the USA are estimated
15 prospective cohorts including 65,064 patients with to be attributable to air pollution38. As the air quality and
incident CKD showed that current smoking is an inde- extent of environmental pollution is far worse in most
pendent risk factor for incident CKD (OR 1.34, 95% CI developing countries than in the USA, air and/or envi-
1.23–1.47), compared with never smokers173. Although ronmental pollution may have a substantial role in the
the prevalence of smoking is decreasing in adults, up aetiology of CKDu in developing countries. It is crucial
to 20–40% of children are still exposed to a living envi- to raise awareness among policy makers, industry repre-
ronment that contains secondhand smoke174,175. In chil- sentatives and the public in developing countries about
dren, exposure to secondhand smoke is associated with environmental pollutants as a public health threat. Many
increased blood pressure, dyslipidaemia176, increased of the strategies for environmental protection that have
C‑reactive protein levels and endothelial dysfunction177. been successfully applied in developed countries, such as
A study reported an association between exposure to establishing and strictly enforcing air quality standards,
second­hand smoke and nephrotic-range proteinuria in mandating vehicle and industrial emissions controls,
children with mild to moderate CKD, even after a­ djusting adopting policies against heavily polluting industries,
for other known risk factors178. promoting the use of greener energy and public trans-
portation and prioritizing clinical and health-care
Chronic kidney disease of unknown aetiology. Diabetes research into the effects of pollutants, could also be
and hypertension are the leading causes of CKD in devel- applied in developing countries.
oped countries, whereas glomerulonephritis remains Most of the epidemiological evidence linking envi-
the main cause of CKD in the developing world179. In the ronmental pollution to kidney diseases discussed here
past two decades, outbreaks of CKD of unknown aeti- are from cross-sectional studies. To establish a causal
ology (CKDu) have been reported in several developing relationship between exposure to environment pol-
countries, including Sri Lanka, India, Egypt and sev- lutants and kidney disease, longitudinal studies with
eral countries in Central America180. Patients with CKDu specific and quantified measurement of environmental
are mainly young male farmers, and the clinical mani­ exposures are required. It is important to investigate the
festations of CKDu are similar to those of interstitial toxic effects of each constituent of various sources of
nephritis. The major histological lesions in the biopsy pollution and to establish the dose–response relation-
samples from patients with CKDu include interstitial ship between a pollutant and kidney disease for a wide
fibrosis, tubular atrophy and interstitial mononuclear range of exposure levels. Most studies on the pathogenic
cell infiltration. Environmental pollutants in contami- mechanisms of pollutants focus on their effects on gen-
nated water and food, including heavy metals, indus- eral pathways, such as systemic inflammation and oxida-
trial chemicals, fertilizers and pesticides, are suspected tive stress, and study of the detailed mechanisms of the
potential causes of CKDu179. In a study carried out in pathogenesis of specific kidney diseases is particularly
Sri Lanka, exposure to glyphosate was associated with lacking. In addition, studying the genetic background of
substantially increased risk of CKDu among male farm- patients and the interaction between environment pol-
workers and residents who drank water polluted with lutants and genetic factors may provide valuable insights
glyphosate181. To identify the aetiologies of CKDu, the into disease susceptibility. Finally, data from high-quality
design of consistent and comparative multisite studies population-based studies will inform regulatory strat­
across high-risk populations might help to elucidate the egies for the control of pollution and reduce or prevent
importance of region-specific versus global risk factors. the exposure of people to environmental health risks.

1. Mills, K. T. et al. A systematic analysis of worldwide estimates. Am. J. Kidney Dis. 62, 253–260 9. Aaron, J. C. et al. Estimates and 25‑year trends of the
population-based data on the global burden of chronic (2013). global burden of disease attributable to ambient air
kidney disease in 2010. Kidney Int. 88, 950–957 5. Brück, K. et al. CKD prevalence varies across the pollution: an analysis of data from the Global Burden
(2015). european general population. J. Am. Soc. Nephrol. of Diseases Study 2015. Lancet 389, 1907–1918
2. GBD 2015 Mortality and Causes of Death 27, 2135–2147 (2016). (2017).
Collaborators. Global, regional, and national life 6. Xie, Y. et al. Long-term kidney outcomes among users 10. Institute for Health Metrics and Evaluation. GBD
expectancy, all-cause mortality, and cause-specific of proton pump inhibitors without intervening acute compare. IHME https://vizhub.healthdata.org/gbd-
mortality for 249 causes of death, 1980–2015: kidney injury. Kidney Int. 91, 1482–1494 (2017). compare (2016).
a systematic analysis for the Global Burden of 7. Prüss-Ustün, A., Wolf, J., Corvalán, C., Bos, R. & 11. Brook, R. D. et al. Air pollution and cardiovascular
Disease Study 2015. Lancet 388, 1459–1544 Neira, M. Preventing disease through healthy disease: a statement for healthcare professionals from
(2016). environments: a global assessment of the burden the Expert Panel on Population and Prevention
3. Coresh, J. et al. Prevalence of chronic kidney of disease from environmental risks (World Health Science of the American Heart Association. Circulation
disease in the United States. JAMA 298, 2038–2047 Organization, 2016). 109, 2655–2671 (2004).
(2007). 8. Brook, R. D. et al. Particulate matter air pollution and 12. Carlsten, C. et al. Diesel exhaust augments allergen-
4. Grams, M. E., Juraschek, S. P. & Selvin, E. Trends in cardiovascular disease: an update to the scientific induced lower airway inflammation in allergic
the prevalence of reduced GFR in the United States: statement from the American Heart Association. individuals: a controlled human exposure study.
a  comparison of creatinine- and cystatin C‑based Circulation 121, 2331–2378 (2010). Thorax 71, 35–44 (2016).

NATURE REVIEWS | NEPHROLOGY VOLUME 14 | MAY 2018 | 321


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

13. Liu, Y. et al. Air pollution and adverse cardiac 39. Donaldson, K. et al. Combustion-derived 64. Zheng, Y. & Ayotte, J. D. At the crossroads: hazard
remodeling: clinical effects and basic mechanisms. nanoparticles: a review of their toxicology following assessment and reduction of health risks from arsenic
Front. Physiol. 6, 162 (2015). inhalation exposure. Part. Fibre Toxicol. 2, 10 (2005). in private well waters of the northeastern United
14. Kelly, F. J. et al. Monitoring air pollution: use of early 40. Carvalho, T. C. & Peters, J. I. and Williams 3rd, R. O. States and Atlantic Canada. Sci. Total Environ. 505,
warning systems for public health. Respirology 17, Influence of particle size on regional lung deposition- 1237–1247 (2015).
7–19 (2012). what evidence is there? Int. J. Pharm. 406, 1–10 65. Brouwer, R. et al. Comparing willingness to pay for
15. Kellogg, W. W. et al. The sulfur cycle. Science 175, (2011). improved drinking-water quality using stated
587–596 (1972). 41. Nemmar, A. et al. Diesel exhaust particles in the lung preference methods in rural and urban Kenya.
16. Bruce, N., Perez-Padilla, R. & Albalak, R. Indoor air aggravate experimental acute renal failure. Toxicol. Sci. Appl. Health Econ. Health Policy 13, 81–94 (2015).
pollution in developing countries: a major 113, 267–277 (2010). 66. Fox, D. I. et al. Removing heavy metals in water: the
environmental and public health challenge. 42. Waly, M. I., Ali, B. H. & Nemmar, A. Acute effects of interaction of cactus mucilage and arsenate (As (V)).
Bull. World Health Organ. 78, 1078–1092 (2000). diesel exhaust particles and cisplatin on oxidative Environ. Sci. Technol. 46, 4553–4559 (2012).
17. Larkin, A. et al. Relationships between changes in stress in cultured human kidney (HEK 293) cells, and 67. Wolf, J., Bonjour, S. & Prüss-Ustün, A. An exploration
urban characteristics and air quality in East Asia from the influence of curcumin thereon. Toxicol. In Vitro 27, of multilevel modeling for estimating access to
2000 to 2010. Environ. Sci. Technol. 50, 9142–9149 2299–2304 (2013). drinking-water and sanitation. J. Water Health 11,
(2016). 43. Nemmar, A. et al. Potentiation of cisplatin-induced 64–77 (2013).
18. [No authors listed.] Ambient (outdoor) air pollution in nephrotoxicity by repeated exposure to diesel exhaust 68. Onda, K., LoBuglio, J. & Bartram, J. Global access
cities database 2014. World Health Organization particles: An experimental study in rats. Exp. Biol. to safe water: accounting for water quality and the
http://www.who.int/phe/health_topics/outdoorair/ Med. 239, 1036–1044 (2014). resulting impact on MDG progress. Int. J. Environ. Res.
databases/cities-2014/en/ (2014). 44. Nemmar, A. et al. Prolonged pulmonary exposure to Publ. Health 9, 880–894 (2012).
19. Xu, X. et al. Long-term exposure to air pollution and diesel exhaust particles exacerbates renal oxidative 69. Satpathy, D., Reddy, M. V. & Dhal, S. P. Risk
increased risk of membranous nephropathy in China. stress, inflammation and dna damage in mice with assessment of heavy metals contamination in paddy
J. Am. Soc. Nephrol. 27, 3739–3746 (2016). adenine-induced chronic renal failure. Cell. Physiol. soil, plants, and grains (Oryza sativa L.) at the East
20. Dockery, D. W. & Pope, C. A. 3rd. Acute respiratory Biochem. 38, 1703–1713 (2016). Coast of India. Biomed. Res. Int. 2014, 545473
effects of particulate air pollution. Annu. Rev. Public 45. Al Suleimani, Y. M. et al. Effect of diesel exhaust (2014).
Health 15, 107–132 (1994). particles on renal vascular responses in rats with 70. Wu, Q. et al. Heavy metal contamination of soil and
21. Holder, A. L. et al. Inflammatory response of lung chronic kidney disease. Environ. Toxicol. 32, 541–549 water in the vicinity of an abandoned e‑waste recycling
cells exposed to whole, filtered, and hydrocarbon (2016). site: implications for dissemination of heavy metals.
denuded diesel exhaust. Chemosphere 70, 13–19 46. Ali, B. H. et al. The effect of thymoquinone treatment Sci. Total Environ. 507, 217–225 (2015).
(2007). on the combined renal and pulmonary toxicity of 71. Arnich, N. et al. Dietary exposure to trace elements
22. Heeb, N. V. et al. PCDD/F formation in an iron/ cisplatin and diesel exhaust particles. Exp. Biol. Med. and health risk assessment in the 2nd French Total
potassium-catalyzed diesel particle filter. Environ. Sci. 240, 1698–1707 (2015). Diet Study. Food Chem. Toxicol. 50, 2432–2449
Technol. 47, 6510–6517 (2013). 47. Atkinson, R. W. et al. Acute effects of particulate air (2012).
23. Jayaram, V. et al. Real-time gaseous, PM and ultrafine pollution on respiratory admissions: results from 72. Mortensen, M. E., Wong, L. Y. & Osterloh, J. D.
particle emissions from a modern marine engine APHEA 2 project. Am. J. Respir. Crit. Care Med. 164, Smoking status and urine cadmium above levels
operating on biodiesel. Environ. Sci. Technol. 45, 1860–1866 (2001). associated with subclinical renal effects in U. S. adults
2286–2292 (2011). 48. Thomson, E. M. et al. Ozone inhalation provokes without chronic kidney disease. Int. J. Hyg. Environ.
24. Bahadar, H., Mostafalou, S. & Abdollahi, M. Current glucocorticoid-dependent and -independent effects on Health 214, 305–310 (2011).
understandings and perspectives on non-cancer health inflammatory and metabolic pathways. Toxicol. Sci. 73. Jarup, L. et al. Biological half-time of cadmium in the
effects of benzene: a global concern. Toxicol. Appl. 152, 17–28 (2016). blood of workers after cessation of exposure.
Pharmacol. 276, 83–94 (2014). 49. Thomson, E. M. et al. Mapping acute systemic effects of Scand. J. Work. Environ. Health 9, 327–331 (1983).
25. Wang, Z. L. Association between chronic obstructive inhaled particulate matter and ozone: multiorgan gene 74. Satarug, S. et al. Modeling cadmium exposures in low-
pulmonary disease and lung cancer: the missing link. expression and glucocorticoid activity. Toxicol. Sci. 135, and high-exposure areas in Thailand. Environ. Health
Chin. Med. J. 126, 154–165 (2013). 169–181 (2013). Perspect. 121, 531–536 (2013).
26. Kampa, M. & Castanas, E. Human health effects of air 50. Ponticelli, C. & Glassock, R. J. Glomerular diseases: 75. Tsukahara, T. et al. Rice as the most influential source
pollution. Environ. Poll. 151, 362–367 (2008). membranous nephropathy‑a modern view. Clin. J. Am. of cadmium intake among general Japanese
27. U.S. Environmental Protection Agency. Integrated Soc. Nephrol. 9, 609–616 (2014). population. Sci. Total Environ. 305, 41–51 (2003).
Science Assessment (ISA) for Oxides of Nitrogen — 51. Granata, F. et al. Activation of cytokine production 76. Akesson, A. et al. Tubular and glomerular kidney
Health Criteria (Final Report, 2016). EPA https://cfpub. by secreted phospholipase A2 in human lung effects in Swedish women with low environmental
epa.gov/ncea/isa/recordisplay.cfm?deid=310879 macrophages expressing the M‑type receptor. cadmium exposure. Environ. Health Perspect. 113,
(2016). J. Immunol. 174, 464–474 (2005). 1627–1631 (2005).
28. Xing, Y. F., Xu, Y. H., Shi, M. H. & Lian, Y. X. The impact 52. Beck Jr, L. H. et al. M‑Type phospholipase A2 receptor 77. Chaumont, A. et al. Associations between proteins and
of PM2·5 on the human respiratory system. J. Thorac. as target antigen in idiopathic membranous heavy metals in urine at low environmental exposures:
Dis. 8, E69–E74 (2016). nephropathy. N. Engl. J. Med. 361, 11–21 (2009). evidence of reverse causality. Toxicol. Lett. 210,
29. Huang, S. K., Zhang, Q., Qiu, Z. & Chung, K. F. 53. Pfau, J. C., Brown, J. M. & Holian, A. Silica-exposed 345–352 (2012).
Mechanistic impact of outdoor air pollution on mice generate autoantibodies to apoptotic cells. 78. Satarug, S., Vesey, D. A. & Gobe, G. C. Health risk
asthma and allergic diseases. J. Thorac. Dis. 7, Toxicology 195, 167–176 (2004). assessment of dietary cadmium intake: Do current
23–33 (2015). 54. Thompson, A. M. et al. Baseline repeated measures guidelines indicate how much is safe? Environ. Health.
30. Zhang, Q., Qiu, Z., Chung, K. F. & Huang, S. K. Link from controlled human exposure studies: associations Perspect. 125, 284–288 (2017).
between environmental air pollution and allergic between ambient air pollution exposure and the 79. Kobayashi, E. et al. Influence of consumption of
asthma: east meets west. J. Thorac. Dis. 7, 14–22 systemic inflammatory biomarkers IL‑6 and fibrinogen. cadmium-polluted rice or Jinzu River water on
(2015). Environ. Health Perspect. 118, 120–124 (2010). occurrence of renal tubular dysfunction and/or Itai-itai
31. Rhoden, C. R., Wellenius, G. A., Ghelfi, E., Lawrence, J. 55. Stanescu, H. C. et al. Risk HLA‑DQA1 and PLA2R1 disease. Biol. Trace Elem. Res. 127, 257–268 (2009).
& Gonzalez-Flecha, B. PM‑induced cardiac oxidative alleles in idiopathic membranous nephropathy. 80. Nordberg, G. F. Historical perspectives on cadmium
stress and dysfunction are mediated by autonomic N. Engl. J. Med. 364, 616–626 (2011). toxicology. Toxicol. Appl. Pharmacol. 238, 192–200
stimulation. Biochim. Biophys. Acta 1725, 305–313 56. Cui, Z. et al. MHC class II risk alleles and amino acid (2009).
(2005). residues in idiopathic membranous nephropathy. 81. Templeton, D. M. & Chaitu, N. Effects of divalent
32. Hesterberg, T. W. et al. Non-cancer health effects of J. Am. Soc. Nephrol. 28, 1651–1664 (2017). metals on the isolated rat glomerulus. Toxicology 61,
diesel exhaust: a critical assessment of recent human 57. Le, W. B. et al. HLA‑DRB1*15:01 and 119–133 (1990).
and animal toxicological literature. Crit. Rev. Toxicol. HLA‑DRB3*02:02 in PLA2R‑related membranous 82. Dorian, C., Gattone 2nd, V. H. & Klaasen, C. D.
39, 195–227 (2009). nephropathy. J. Am. Soc. Nephrol. 28, 1642–1650 Renal cadmium deposition and injury as a result of
33. Carlisle, A. J. & Sharp, N. C. Exercise and outdoor (2017). accumulation of cadmium-metallothionein (CdMT) by
ambient air pollution. Br. J. Sports Med. 35, 58. Järup, L. Hazards of heavy metal contamination. Br. the proximal convoluted tubules — alight microscopic
214–222 (2001). Med. Bull. 68,167–182 (2003). autoradiography study with 109CdMT. Toxicol. Appl.
34. Ristovski, Z. D. et al. Respiratory health effects of 59. Cheng, S. Heavy metal pollution in China: origin, Pharmacol. 114, 173–181 (1992).
diesel particulate matter. Respirology 17, 201–212 pattern and control. Environ. Sci. Pollut. Res. Int. 10, 83. Lauwerys, R. R. et al. Characterization of cadmium
(2012). 192–198 (2003). proteinuria in man and rat. Environ. Health Perspect.
35. Miller, M. R. et al. Inhaled nanoparticles accumulate 60. Liu, P. et al. Analysis of heavy metal sources for 54, 147–152 (1984).
at sites of vascular disease. ACS Nano 11, vegetable soils from Shandong Province, China. 84. Bernard, A., Lauwerys, R. & Gengoux, P.
4542–4552 (2017). Agric. Sci. China 10, 109–119 (2011). Characterization of the proteinuria induced by
36. Lue, S. H. et al. Residential proximity to major 61. Cheng, Z. Q. et al. Speciation of heavy metals in prolonged oral administration of cadmium in female
roadways and renal function. J. Epidemiol. Commun. garden soils: evidences from selective and sequential rats. Toxicology 20, 345–357 (1981).
Health 67, 629–634 (2013). chemical leaching. J. Soils Sediments 11, 628–638 85. Nair, A. R. et al. Glutathione and mitochondria
37. Mehta, A. J. et al. Long-term exposure to ambient (2011). determine acute defense responses and adaptive
fine particulate matter and renal function in older 62. Zhang, X. et al. Impact of soil heavy metal pollution on processes in cadmium-induced oxidative stress and
men: the Veterans Administration Normative Aging food safety in China. PLoS ONE 10, e0135182 (2015). toxicity of the kidney. Arch. Toxicol. 89, 2273–2289
Study. Environ. Health Perspect. 124, 1353–1360 63. Zeng, G. M. et al. Efficiency of biochar and compost (or (2015).
(2016). composting) combined amendments for reducing Cd, 86. Bork, U. et al. Cadmium-induced DNA damage triggers
38. Bowe, B. et al. Particulate matter air pollution and Cu, Zn and Pb bioavailability, mobility and ecological G2/M arrest via chk1/2 and cdc2 in p53‑deficient
the risk of uncident CKD and progression to ESRD. risk in wetland soil. RSC Adv. 5, 34541–34548 kidney proximal tubule cells. Am. J. Physiol. Renal
J. Am. Soc. Nephrol. 29, 218–230 (2017). (2015). Physiol. 298, 255–265 (2010).

322 | MAY 2018 | VOLUME 14 www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

87. Kim, H. R. et al. Transcriptional regulation, and Nutrition Examination Survey. Arch. Intern. Med. elimination of inorganic mercury mediated by
stabilization, and subcellular redistribution of 170, 75–82 (2010). 2,3‑dimercaptopropane‑1‑sulfonic acid and meso‑2,3-
multidrug resistance-associated protein 1 (MRP1) by 111. Navas-Acien, A. et al. Blood cadmium and lead and dimercaptosuccinic acid. J. Pharmacol. Exp. Ther.
glycogen synthase kinase 3αβ: novel insights on modes chronic kidney disease in US adults: a joint analysis. 324, 383–390 (2008).
of cadmium-induced cell death stimulated by MRP1. Am. J. Epidemiol. 170, 1156–1164 (2009). 138. Zalups, R. K., Aslamkhan, A. G. & Ahmad, S. Human
Arch. Toxicol. 89, 1271–1284 (2015). 112. Cramer, K. et al. Renal ultrastructure, renal function, organic aniontransporter 1 mediates cellular uptake of
88. Mahaffey, K. R., Clickner, R. P. & Bodurow, C. C. Blood and parameters of lead toxicity in workers with cysteine‑S conjugates of inorganic mercury. Kidney Int.
organic mercury and dietary mercury intake: National different periods of lead exposure. Br. J. Ind. Med. 31, 66, 251–261 (2004).
Health and Nutrition Examination Survey, 1999 and 113–127 (1974). 139. Zalups, R. K. & Ahmad, S. Homocysteine and the renal
2000. Environ. Health Perspect. 112, 562–570 113. Goyer, R. A. Mechanisms of lead and cadmium epithelial transport and toxicity of inorganic mercury:
(2004). nephrotoxicity. Toxicol. Lett. 46, 153–162 (1989). role of basolateral transporter organic anion
89. Iesato, K. et al. Renal tubular dysfunction in Minamata 114. Evans, M. & Elinder, C. G. Chronic renal failure from transporter 1. J. Am. Soc. Nephrol. 15, 2023–2031
disease. Detection of renal tubular antigen and lead: myth or evidence-based fact? Kidney Int. 79, (2004).
beta‑2‑microglobin in the urine. Ann. Intern. Med. 86, 272–279 (2011). 140. Aleksunes, L. M. et al. Renal xenobiotic transporters
731–737 (1977). 115. Oyagbemi, A. A. et al. Lack of reversal of oxidative are differentially expressed in mice following cisplatin
90. Bohets, H. H. et al. Cytotoxicity of mercury compounds damage in renal tissues of lead acetate-treated rats. treatment. Toxicology 250, 82–88 (2008).
in LLC‑PK1, MDCK and human proximal tubular cells. Environ. Toxicol. 30, 1235–1243 (2015). 141. Kala, S. V. et al. Formation and urinary excretion of
Kidney Int. 47, 395–403 (1995). 116. Kwon, S. Y. et al. Erythrophagocytosis of lead-exposed arsenic triglutathione and methylarsenic diglutathione.
91. Nakazawa, N., Makino, F. & Okada, S. Acute effects of erythrocytes by renal tubular cells: possible role in Chem. Res. Toxicol. 17, 243–249 (2004).
mercuric compounds on cultured mammalian cells. lead-induced nephrotoxicity. Environ. Health Perspect. 142. Scott, N. et al. Reactions of arsenic(III) and arsenic(V)
Biochem. Pharmacol. 24, 489–493 (1975). 123, 120–127 (2015). species with glutathione. Chem. Res. Toxicol. 6,
92. Sarmad, S. & German, T. H. Environmental exposures, 117. Leonard, A. & Lauwerys, R. R. Carcinogenicity, 102–106 (1993).
socioeconomics, disparities, and the kidneys. teratogenicity and mutagenicity of arsenic. Mutat. Res. 143. Drobna, Z. et al. Metabolism of arsenic in human liver:
Adv. Chron. Kidney Dis. 22, 39–45 (2015). 75, 49–62 (1980). the role of membrane transporters. Arch. Toxicol. 84,
93. Miller, S., Pallan, S., Gangji, A. S., Lukic, D. & 118. Pierce, B. L. et al. Genome-wide association study 3–16 (2010).
Clase, C. M. Mercury-associated nephrotic syndrome: identifies chromosome 10q24.32 variants associated 144. Djukanovic, L. R. & Radovanovic, Z. in Clinical
a case report and systematic review of the literature. with arsenic metabolism and toxicity phenotypes in Nephrotoxins (eds de Broe, M. E., Porter, G. A.,
Am. J. Kidney Dis. 62, 135–138 (2013). Bangladesh. PLoS Genet. 8, e1002522 (2012). Bennett, W. M., Verpooten & G. A.) 588–601 (Kluwer,
94. Li, S. J. et al. Mercury-induced membranous 119. Hughes, M. F. et al. Arsenic exposure and toxicology: 2003).
nephropathy: clinical and pathological features. a historical perspective. Toxicol. Sci. 123, 305–332 145. Petronic, V. in Endemic Nephropathy (eds
Clin. J. Am. Soc. Nephrol. 5, 439–444 (2010). (2011). Radovanovic, Z, Sindjic, M, Polenakovic, M,
95. Chakera, A., Lasserson, D., Beck Jr, L. H., Roberts, I. S. 120. Liu, J. & Waalkes, M. P. Liver is a target of arsenic Djukanovic, L. J. & Petronic, V.) 350–439 (Institute for
& Winearls, C. G. Membranous nephropathy after use carcinogenesis. Toxicol. Sci. 105, 24–32 (2008). Textbook Publishing, Belgrade, 2000).
of UK‑manufactured skin creams containing mercury. 121. Yoshida, T., Yamauchi, H. & Fan Sun, G. Chronic health 146. Grollman, A.P. & Jelakovic, B. Role of environmental
QJM 104, 893–896 (2011). effects in people exposed to arsenic via the drinking toxins in endemic (Balkan) nephropathy. October
96. Hua, J., Pelletier, L., Berlin, M. & Druet, P. water: dose-response relationships in review. 2006, Zagreb, Croatia. J. Am. Soc. Nephrol. 18,
Autoimmune glomerulonephritis induced by mercury Toxicol. Appl. Pharmacol. 198, 243–252 (2004). 2817–2823 (2007).
vapour exposure in the Brown Norway rat. Toxicology 122. Meliker, J. R. et al. Arsenic in drinking water and 147. Vanherweghem, J. L. et al. Rapidly progressive
79, 119–129 (1993). cerebrovascular disease, diabetes mellitus, and kidney interstitial renal fibrosis in young women: association
97. Abedi-Valugerdi, M., Hu, H. & Möller, G. Mercury- disease in Michigan: a standardized mortality ratio with slimming regimen including Chinese herbs.
induced renal immune complex deposits in young analysis. Environ. Health 6, 4 (2007). Lancet 341, 387–391 (1993).
(NZB x NZW)F1 mice: characterization of antibodies/ 123. Jomova, K. & Valko, M. Advances in metal-induced 148. Stiborova, M., Arlt, V. M. & Schmeiser, H. H. Balkan
autoantibodies. Clin. Exp. Immunol. 110, 86–91 oxidative stress and human disease. Toxicology 283, endemic nephropathy: an update on its aetiology.
(1997). 65–87 (2011). Arch. Toxicol. 90, 2595–2615 (2016).
98. Icard, P. et al. Evidence for a role of antilaminin- 124. Chen, Z. et al. Identification of target organs of copper 149. Zeng, Y. et al. Aristolochic acid I induced autophagy
producing B cell clones that escape tolerance in the nanoparticles with ICP‑MS technique. J. Radioanal. extenuates cell apoptosis via ERK 1/2 pathway in
pathogenesis of HgCl2‑induced membranous Nucl. Chem. 272, 599–603 (2007). renal tubular epithelial cells. PLoS ONE 7, e30312
glomerulopathy. Nephrol. Dial. Transplant. 8, 125. Lei, R. et al. Integrated metabolomic analysis of the (2012).
122–127 (1993). nano-sized copper particle-induced hepatotoxicity and 150. Yang, C. C. et al. Autophagy induction promotes
99. Abedi-Valugerdi, M. & Möller, G. Contribution of H-2 nephrotoxicity in rats: a rapid in vivo screening aristolochic acid‑I‑induced renal injury in vivo and
and non‑H-2 genes in the control of mercury-induced method for nanotoxicity. Toxicol. Appl. Pharmacol. in vitro. Toxicology 312, 63–73 (2013).
autoimmunity. Int. Immunol. 12, 1425–1430 232, 292–301 (2008). 151. Zeng, Y. et al. Autophagy inhibitors promoted
(2000). 126. Kumar, V. et al. Relationship of antioxidant and aristolochic acid I induced renal tubular epithelial cell
100. Pollard, K. M., Hultman, P. & Kono, D. H. Immunology oxidative stress markers in different organs following apoptosis via mitochondrial pathway but alleviated
and genetics of induced systemic autoimmunity. copper toxicity in a rat model. Toxicol. Appl. Pharmacol. nonapoptotic cell death in mouse acute aritolochic
Autoimmun. Rev. 4, 282–288 (2005). 293, 37–43 (2016). acid nephropathy model. Apoptosis 19, 1215–1224
101. Gardner, R. M. et al. Mercury exposure, serum 127. Wu, B. Y. et al. Toxicological effects of dietary nickel (2014).
antinuclear/antinucleolar antibodies, and serum chloride on intestinal microbiota. Ecotoxicol. Environ. 152. Grollman, A. P. et al. Aristolochic acid and the etiology
cytokine levels in mining populations in Amazonian Safety 109, 70–76 (2014). of endemic (Balkan) nephropathy. Proc. Natl Acad. Sci.
Brazil: a cross-sectional study. Environ. Res. 110, 128. Pasanen, K. et al. Mortality among population with USA 104, 12129–12134 (2007).
345–354 (2010). exposure to industrial air pollution containing nickel 153. Hoang, M. L. et al. Mutational signature of aristolochic
102. Motts, J. A., Shirley, D. L., Silbergeld, E. K. & and other toxic metals. J. Occup. Environ. Med. 54, acid exposure as revealed by whole-exome
Nyland, J. F. Novel biomarkers of mercury-induced 583–591. sequencing. Sci. Transl Med. 5, 197ra102 (2013).
autoimmune dysfunction: a cross-sectional study in 129. Guo, H. et al. Modulation of the PI3K/Akt pathway 154. Jelakovic, B. et al. Aristolactam–DNA adducts are a
Amazonian Brazil. Environ. Res. 132, 12–18 (2014). and Bcl‑2 family proteins involved in chicken’s tubular biomarker of environmental exposure to aristolochic
103. Jedrychowski, W. et al. Very low prenatal exposure to apoptosis induced by nickel chloride (NiCl2). acid. Kidney Int. 81, 559–567 (2012).
lead and mental development of children in infancy Int. J. Mol. Sci. 16, 22989–23011 (2015). 155. Moriya, M. et al. TP53 Mutational signature for
and early childhood: Krakow prospective cohort study. 130. Guo, H. et al. Nickel chloride (NiCl2)-caused aristolochic acid: an environmental carcinogen.
Neuroepidemiology 32, 270–278 (2009). inflammatory responses via activation of NF‑κB pathway Int. J. Cancer 129, 1532–1536 (2011).
104. Bellinger, D. C. Very low lead exposures and children’s and reduction of anti-inflammatory mediator expression 156. Poon, S. L. et al. Genome-wide mutational signatures
neurodevelopment. Curr. Opin. Pediatr. 20, 172–177 in the kidney. Oncotarget 6, 28607–28620 (2015). of aristolochic acid and its application as a screening
(2008). 131. Fujishiro, H. et al. Roles of ZIP8, ZIP14, and DMT1 tool. Sci. Transl Med. 5, 197ra101 (2013).
105. Liu, J. et al. Prenatal and postnatal lead exposure and in transport of cadmium and manganese in mouse 157. National Toxicology Program. Toxicology and
cognitive development of infants followed over the first kidney proximal tubule cells. Metallomics 4, 700–708 carcinogenesis studies of ochratoxin A (CAS No.
three years of life: a prospective birth study in the (2014). 303‑47‑9) in F344/N Rats (Gavage Studies).
Pearl River Delta region, China. Neurotoxicology 44, 132. Lai, L. H. et al. Renal dysfunction and hyperuricemia Natl Toxicol. Program Tech. Rep. Ser. 358, 1–142
326–334 (2014). with low blood lead levels and ethnicity in community- (1989).
106. Shah-Kulkarni, S. et al. Neurodevelopment in early based study. Sci. Total Environ. 401, 39–43 (2008). 158. Fuchs, R. & Peraica, M. Ochratoxin A in human kidney
childhood affected by prenatal lead exposure and iron 133. Goyer, R. A. Toxic and essential metal interactions. diseases. Food Addit. Contam. 22, 53–57 (2005).
intake. Medicine 95, e2508 (2016). Annu. Rev. Nutr. 17, 37–50 (1997). 159. Wafa, E. et al. Human ochratoxicosis and nephropathy
107. Hara, A. et al. Incidence of nephrolithiasis in relation 134. Bogden, J. D. et al. Dietary calcium modifies in Egypt: a preliminary study. Hum. Exp. Toxicol. 17,
to environmental exposure to lead and cadmium in a concentrations of lead and other metals and renal 124–129 (1998).
population study. Environ. Res. 145, 1–8 (2016). calbindin in rats. J. Nutr. 122, 1351–1360 (1992). 160. Malir, F. et al. Ochratoxin A: 50 years of research.
108. Kim, N. H. et al. Environmental heavy metal exposure 135. Blake, K. C. & Mann, M. Effect of calcium and Toxins 8, 191(2016).
and chronic kidney disease in the general population. phosphorus on the gastrointestinal absorption of 161. Kim, K. H., Kabir, E. & Jahan, S. A. Exposure to
J. Kor. Med. Sci. 30, 272–277 (2015). 203Pb in man. Environ. Res. 30, 188–194 (1983). pesticides and the associated human health effects.
109. Garcia-Esquinas, E. et al. Association of lead and 136. Li, S. J. et al. Mercury-induced membranous Sci. Total Environ. 575, 525–535 (2017).
cadmium exposure with frailty in US older adults. nephropathy: clinical and pathological features. 162. Kim, S. J., Gil, H. W., Yang, J. O., Lee, E. Y. &
Environ. Res. 137, 424–431 (2015). Clin. J. Am. Soc. Nephrol. 5, 439–444 (2010). Hong, S. Y. The clinical features of acute kidney
110. Fadrowski, J. J. et al. Blood lead level and kidney 137. Bridges, C. C., Joshee, L. & Zalups, R. K. injury in patients with acute paraquat intoxication.
function in US adolescents: the Third National Health Multi drug resistance proteins and the renal Nephrol. Dial. Transplant. 24, 1226–1232 (2009).

NATURE REVIEWS | NEPHROLOGY VOLUME 14 | MAY 2018 | 323


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

163. Mohamed, F. et al. Mechanism-specific injury 171. Hau, A. K., Kwan, T. H. & Li, P. K. Melamine toxicity and 180. Lunyera, J. et al. CKD of uncertain etiology:
biomarkers predict nephrotoxicity early following the kidney. J. Am. Soc. Nephrol. 20, 245–250 (2009). a  systematic review. Clin. J. Am. Soc. Nephrol. 11,
glyphosate surfactant herbicide (GPSH) poisoning. 172. Yang, L. et al. Four years follow‑up of 101 children with 379–385 (2016).
Toxicol. Lett. 258, 1–10 (2016). melamine-related urinary stones. Urolithiasis 41, 181. Jayasumana, C. et al. Drinking well water and
164. Sonne, C. et al. Organochlorine-induced 265–266 (2013). occupational exposure to herbicides is associated with
histopathology in kidney and liver tissue from Arctic 173. Xia, J. et al. Cigarette smoking and chronic kidney chronic kidney disease, in Padavi-Sripura, Sri Lanka.
fox (Vulpes lagopus). Chemosphere 71, 1214–1224 disease in the general population: a systematic review Environ. Health 14, 6 (2015).
(2008). and meta-analysis of prospective cohort studies.
165. Adachi, J. et al. 7‑Hydroperoxycholesterol as a marker Nephrol. Dial. Transplant. 32, 475–487(2017). Acknowledgements
of oxidative stress in rat kidney induced by paraquat. 174. Omoloja, A. & Tyc, V. L. Tobacco and the pediatric The authors are supported by the National Key Technology
Free Radic. Res. 33, 321–327 (2000). chronic kidney disease population. Pediatr. Nephrol. S u p p o r t P ro g ra m o f C h i n a ( 2 01 3 BA I 0 9 B 0 6 a n d
166. Lebov, J. F. et al. Pesticide exposure and end-stage 30, 235–243 (2015). 2015BAI12B07 to F.F.H.), the National Natural Science
renal disease risk among wives of pesticide applicators 175. Omoloja, A. et al. Tobacco exposure in children and Foundation of China (81770683 to X.X.), the National
in the agricultural health study. Environ. Res. 143, adolescents with chronic kidney disease: parental Natural Science Foundation of China (Key Program)
198–210 (2015). behavior and knowledge. A study from the Midwest (81430016 to F.F.H.), the Major Scientific and Technological
167. Lebov, J. F. et al. Pesticide use and risk of end-stage Pediatric Nephrology Consortium. Clin. Nephrol. 81, Planning Project of Guangzhou (201504010027 to F.F.H.)
renal disease among licensed pesticide applicators in 307–312 (2014). and the Major International (Regional) Joint Research Project
the agricultural health study. Occup. Environ. Med. 176. Neufeld, E. J., Mietus-Snyder, M., Beiser, A. S., (81620108003 to F.F.H.).
73, 3–12 (2016). Baker, A. L. & Newburger, J. W. Passive cigarette
168. Mesnage, R. et al. Transcriptome profile analysis smoking and reduced HDL cholesterol levels in children Author contributions
reflects rat liver and kidney damage following chronic with high-risk lipid profiles. Circulation 96, 1403–1407 All authors researched data for the article, contributed sub-
ultra-low dose Roundup exposure. Environ. Health 14, (1997). stantially to discussion of the article’s content, wrote the arti-
70 (2015). 177. Kallio, K. et al. Tobacco smoke exposure is associated cle and reviewed and/or edited the manuscript before
169. Siddarth, M. et al. Increased level of organochlorine with attenuated endothelial function in 11‑year-old submission.
pesticides in chronic kidney disease patients of healthy children. Circulation 115, 3205–3212 (2007).
unknown etiology: role of GSTM1/GSTT1 178. Omoloja, A. et al. Secondhand smoke exposure is Competing interests
polymorphism. Chemosphere 96, 174–179 associated with proteinuria in children with chronic The authors declare no competing interests.
(2014). kidney disease. Pediatr. Nephrol. 28, 1243–1251
170. Kataria, A. et al. The effects of environmental (2013). Publisher’s note
chemicals on renal function. Nat. Rev. Nephrol. 11, 179. Levey, A. S. & Coresh, J. Chronic kidney disease. Lancet Springer Nature remains neutral with regard to jurisdictional
610–625 (2015). 379, 165–180 (2012). claims in published maps and institutional affiliations.

324 | MAY 2018 | VOLUME 14 www.nature.com/nrneph


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like