ArticlePDF Available

In Utero and Postnatal Exposure to Arsenic Alters Pulmonary Structure and Function

Authors:
  • Odyssey Research Institute

Abstract and Figures

In addition to cancer endpoints, arsenic exposures can also lead to non-cancerous chronic lung disease. Exposures during sensitive developmental time points can contribute to the adult disease. Using a mouse model, in utero and early postnatal exposures to arsenic (100 ppb or less in drinking water) were found to alter airway reactivity to methacholine challenge in 28 day old pups. Removal of mice from arsenic exposure 28 days after birth did not reverse the alterations in sensitivity to methacholine. In addition, adult mice exposed to similar levels of arsenic in drinking water did not show alterations. Therefore, alterations in airway reactivity were irreversible and specific to exposures during lung development. These functional changes correlated with protein and gene expression changes as well as morphological structural changes around the airways. Arsenic increased the whole lung levels of smooth muscle actin in a dose dependent manner. The level of smooth muscle mass around airways was increased with arsenic exposure, especially around airways smaller than 100 μm in diameter. This increase in smooth muscle was associated with alterations in extracellular matrix (collagen, elastin) expression. This model system demonstrates that in utero and postnatal exposure to environmentally relevant levels of arsenic can irreversibly alter pulmonary structure and function in the adults.
Content may be subject to copyright.
In Utero and Postnatal Exposure to Arsenic Alters Pulmonary
Structure and Function
R. Clark Lantza,b,c,*, Binh Chaua, Priyanka Sarihana, Mark L. Wittenb,d, Vadim I.
Pivniouka,e, and Guan Jie Chena
a Department of Cell Biology and Anatomy, University of Arizona, Tucson, AZ 85724
b Southwest Environmental Health Science Center, University of Arizona, Tucson, AZ 85721
c BIO5 Institute, University of Arizona, Tucson, AZ 85721
d Department of Pediatrics, University of Arizona, Tucson, AZ 85724
e Arizona Respiratory Center, University of Arizona, Tucson, AZ 85724
Abstract
In addition to cancer endpoints, arsenic exposures can also lead to non-cancerous chronic lung
disease. Exposures during sensitive developmental time points can contribute to the adult disease.
Using a mouse model, in utero and early postnatal exposures to arsenic (100 ppb or less in drinking
water) were found to alter airway reactivity to methacholine challenge in 28 day old pups. Removal
of mice from arsenic exposure 28 days after birth did not reverse the alterations in sensitivity to
methacholine. In addition, adult mice exposed to similar levels of arsenic in drinking water did not
show alterations. Therefore, alterations in airway reactivity were irreversible and specific to
exposures during lung development. These functional changes correlated with protein and gene
expression changes as well as morphological structural changes around the airways. Arsenic
increased the whole lung levels of smooth muscle actin in a dose dependent manner. The level of
smooth muscle mass around airways was increased with arsenic exposure, especially around airways
smaller than 100 μm in diameter. This increase in smooth muscle was associated with alterations in
extracellular matrix (collagen, elastin) expression. This model system demonstrates that in utero and
postnatal exposure to environmentally relevant levels of arsenic can irreversibly alter pulmonary
structure and function in the adults.
Keywords
arsenic; lung development; pulmonary function; airway smooth muscle; extracellular matrix
Introduction
Growth and development requires the temporal and spatial coordinated expression of genes
and gene products. During this critical time, in utero and early postnatal exposure to toxicants
* Corresponding author: email – E-mail: lantz@email.arizona.edu, voice – 520-626-6716, FAX – 520-626-2097.
Conflict of Interest: The authors declare that there is no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers
we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting
proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could
affect the content, and all legal disclaimers that apply to the journal pertain.
NIH Public Access
Author Manuscript
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
Published in final edited form as:
Toxicol Appl Pharmacol. 2009 February 15; 235(1): 105–113. doi:10.1016/j.taap.2008.11.012.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
has the potential to affect gene expression, altering organ structure and physiological function
which can be manifested as adult disease (Merkus et al, 2003; Wei et al, 2007). While the
potential adverse health outcomes that result from exposures during these sensitive
developmental times are recognized, only limited attention has been paid to the effects of
environmentally relevant exposures to toxicants during these critical periods of development
(Mazumder, 2007, Vahter, 2008) Inorganic arsenic is a ubiquitous environmental toxicant,
found in high concentrations throughout the world. Chronic environmental arsenic exposure
through consumption of geologically contaminated drinking water has been correlated with
increased incidence of and mortality due to internal cancers of the lung, skin, kidney, urinary
bladder and liver (Chen et al., 1988; Chiou et al., 1995; Wu et al., 1989; Hopenhayn-Rich et
al., 1998). In addition, reports from human studies in Chile, Bangladesh and the West Bengal
region of India show that chronic exposure to arsenic via drinking water is correlated with
increased incidence of chronic cough, chronic bronchitis, shortness of breath and obstructive
or restrictive lung disease (von Ehrenstein, et al., 2005; Mazumder et al., 2000; Smith et al.,
1998). Taken together, these studies argue unequivocally that the lung is targeted by arsenic,
producing both carcinogenic and non-carcinogenic endpoints.
That high exposures to arsenic in drinking water (800 ppb) during sensitive developmental
times can lead to adverse health outcomes and increased mortality has been reported (Smith
et al, 2006). Drinking water exposures to high levels of arsenic either in utero or during early
childhood development led to an increased risk of dying from lung cancers and chronic lung
disease in young adults. Exposures in early childhood led to a standardized mortality ratio
(SMR) for lung cancer of 7.0 and a SMR for bronchiectasis of 12.4. For those exposed both
during in utero and early childhood, the SMRs were 6.1 for lung cancer and 46.2 for
bronchiectasis. These findings suggest that exposure to arsenic in drinking water during early
childhood or in utero has pronounced pulmonary effects, greatly increasing subsequent
mortality in young adults from both malignant and nonmalignant lung disease. The effects and
the molecular targets for alterations after exposure to environmentally relevant levels (0 to 100
ppb) of arsenic, levels that would be seen in some regions of the United States, are not known.
In addition to the effects reported on the lung, early developmental exposures have also been
associated with other adverse outcomes in humans. Arsenic is able to cross the placenta
(Concha et al, 1998). In Chilean populations with well defined arsenic exposures, an association
between arsenic exposure in the drinking water and adverse reproductive outcomes (increase
infant mortality (Hopenhayn-Rich et al, 2000) and decreased birth weight (Hopenhayn et al,
2003)) were suggested. Autopsy tissues from five children living in the Antofagasta area of
Chile (high arsenic exposure region) revealed increased arterial intimal thickening (Rosenberg,
1974). No reports exist concerning the relationship of arsenic exposures and lung function in
children.
Animal and in vitro models have been used in attempts to determine the sites and the
mechanisms of developmental toxicity of inorganic arsenicals. In a mouse model of
transplacental carcinogenesis, arsenic exposure (42.5 and 85 ppm) during gestation days 8
through 18 lead to significant increases in tumor incidence and multiplicity in the lung and
several other organs in adult offspring (Waalkes et al, 2003, Shen et al, 2007). While the doses
used in the previous mouse studies are high compared to environmental exposure levels, they
do show that tumor formation can occur in an animal model of in utero arsenic exposure. There
are also a few studies detailing alterations in the developing fetus induced by maternal arsenic
exposure. Acute, high dose ip injections of arsenic (30-45 mg/kg) during gestation have been
associated with neural tube defects and corresponding aberrant gene expression of
developmentally important transcription factors in the neural tube, including Hox 3.1 and Pax
3 (Liu et al, 2006). These doses of arsenic also triggered upregulation of bcl-2 and p53 gene
expression in the neural tube, indicative of inhibition of cellular proliferation (Wlodarczyk et
Lantz et al. Page 2
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
al, 1996). In our previous research we have exposed pregnant female rats to 500 ppb arsenite
in drinking water from conception until embryonic day 18. Analysis of arsenic-induced
alterations in genes and proteins indicated that targets of in utero arsenic exposure in the
developing lung appear to be the developing extracellular matrix and the processes of cellular
differentiation and branching morphogenesis (Petrick et al, 2008). The most likely affected
pathway was alteration in integrin signaling through the β-catenin pathway, altering c-myc.
Since our earlier research had identified extracellular matrix as a potential target for arsenic,
we hypothesized that arsenic-induced changes in matrix during lung development would lead
to structural and functional alterations in the adult. During fetal and early postnatal lung
development, extracellular matrix gene expression is necessary for proper development of lung
and blood vessels (Mariani et al, 2002). Agents that can alter this expression during these
critical times are known to cause long term morphological alterations in the lung and blood
vessels. (Examples are inhibition of elastin expression following maternal cigarette smoking
(Collins et al, 1985) or postnatal exposure to hyperoxia (Bruce et al, 1996), viral infections
(Castleman et al, 1988) or dexamethasone (Blanco and Frank, 1993)). These exposures lead
to permanent alveolar enlargement. We purpose that arsenic exposure during critical times of
development will result in similar irreversible and morphological changes in the lung. In this
report we present data that show that in utero and early postnatal exposure to environmentally
relevant levels of arsenic in drinking water can lead to irreversible alterations in lung function.
These are accompanied by changes in matrix gene expression and alterations in airway smooth
muscle.
Methods and Materials
Animals and exposure
C57Bl/6 mice were used. For mouse breeding we used non-sterile micro-isolator pan with
water from a sterile double deionized source (used to make arsenic water). We used Teklad
Global 19% protein diet which is good for breeding mice and sani-chip bedding (Harlan Teklad,
Los Angeles, CA). The chow was assessed for total arsenic levels and was found to contain 40
ng/g (40 ppb) of total arsenic. Control animals for this study received distilled water throughout.
Treated animals received sodium arsenite in their drinking water at doses of 5, 10, 50 or 100
μg/l (ppb). These are environmental levels found in drinking water throughout the world. All
solutions were prepared with distilled water and, if necessary, titrated to a pH of 7.0, a pH
similar to that of control water. Each water bottle was placed in a specially made metal casing
to reduce light exposure and subsequent chemical breakdown. The amount of water consumed
by each pen of animals (4 maximum) was monitored and recorded every 24 hours. Bottles were
cleaned and solutions prepared daily. Arsenic speciation was determined and concentrations
verified using ICP-MS, available through the Arizona Superfund Core Facility. Females began
exposure to arsenic in their drinking water two weeks prior to mating. Females used for multiple
breeding cycles were continuously kept on the same arsenic water throughout. Results from
pups born as first litters to breeding females were not different when compared to subsequent
litters. After birth of the pups, mothers and weaned animals were exposed to the arsenic in the
drinking water. Pups were sacrificed using CO2, on day 1, 7, 12 and 28 after birth. During that
time, animals were continually exposed to arsenic in the drinking water. Only one pup from a
litter was used for each time point and measurement (If, for example, N=5, then each of the
five animals used came from a different litter). All protocols were approved by the University
of Arizona, Institutional Animal Care and Use Committee.
Quantitative RT-PCR
All lung tissue was harvested and placed in RNAlater solution from Qiagen Cat. No. 76104 in
ratio of 1ml/100mg of tissue, stored in 4°C overnight. The solution was then removed and
Lantz et al. Page 3
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
samples were stored in -80°C until used. All RNA isolation was done following procedure
using Qiagen RNeasy Mini Kit Cat. No. 74104. We also performed in-column DNAse
treatment (DNAse Kit Cat No. 79254) to remove DNA contamination during the RNA
isolation. 30 mg of tissue per reaction was homogenized with disposable plastic mortar and
pestle.
Lung total RNA was quantified and checked for RNA integrity by Agilent 2100 Bioanalyzer.
cDNA was synthesized using 2 μg of total RNA following procedure from Taqman Reverse
Transcription kit (Applied Biosystems, N808-0234.) Amplification was performed by PCR
using Taq Gold polymerase master mix (Applied Biosystems, N808-0241) and 1× SYBR
Green (Invitrogen, S7563) performed by Roter-gene 3000 (Corbett Research, Australia.). The
PCR conditions were: for collagen 1A1, 1A2, 3A1, smooth muscle actin and beta actin PCR
at 95°C for 30 sec; annealing at 50°C for 30 sec, and extension at 72°C for 30 sec and for elastin
PCR at 95°C for 30 sec; annealing at 58°C for 30sec, and extension at 72°C 30 sec. The PCR
primer sequences are shown in Table 1. Levels of expression were normalized to GAPDH.
After birth, lung GAPDH expression levels do not change
((http://lungtranscriptome.bwh.harvard.edu/)).
Relative quantization of expression levels was obtained by developing a standard curve using
2 μg total RNA from control animals. cDNA was serially diluted and PCR was performed on
these serially diluted cDNAs to obtain the STD curve. For quantifying, the samples from the
control group and treatment groups cDNA were diluted 20 times prior to real-time PCR to
obtain a threshold cycle. The standard curve was then used to obtain fold change in expression.
Immunohistochemistry and lung morphometry
Immunohistochemistry was utilized to localize alpha smooth muscle actin. (Rabbit IgG,
ab5694, Abcam, MA). Lungs were fixed by intratracheal instillation of buffered formalin at a
constant pressure of 20 cm H20. Paraffin embedded sections (5 μm) were baked for 1 hr at 65°
C then subjected to a series of de-paraffinization (3 times of 5 min in xylene) and dehydration.
Antigen retrieval was performed by microwave treatment. The slides were place in 10 mM
citrate buffer at pH 6.0.
All procedures are followed according to Vectorstain ABC kit for Rabbit IgG (PK6101).
Primary antibody staining was with a 1:100 dilution of antibody overnight in a humidity
chamber. After washing, sections were incubated for 30 min with a biotinylated secondary
antibody followed by 30 min incubation with an avidin-HRP complex. Staining was developed
(2 min) using the Vector VIP staining solution (SK4600, Vector Laboratories).
The amount of smooth muscle and collagen around airways was quantitated by analyzing
digital images collected using PCI software (Pittsburgh, PA). Sections of lung tissue were
scanned and all airways cut in cross section (the ratio of maximum to minimum diameter was
less than 2) were analyzed. Diameters were determined by filling of the area inside of the airway
epithelium. PCI is able to obtain the minimum and maximum diameters of a region of interest.
Basement membrane perimeter was determined by tracing. The area of smooth muscle was
determined by thresholding the images to detect only the antibody staining and measuring the
number of pixels detected (Camateros et al, 2007). Area of collagen staining was obtained
using polarized light microscopy of sirius red stained sections (Last et al, 2004). Area of smooth
muscle and collagen staining were then normalized to the square of the basement membrane
perimeter (Camateros et al, 2007). Data were analyzed for all airways and also were subdivided
by airway diameter (small airways, diameter < 100 μm versus large airways, diameter > than
100 μm). The minimum diameter was used a measure of the airway diameter (Weibel, 1979).
Lantz et al. Page 4
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Western Blots
The following antibodies and dilutions were used for Western blot analysis: Procollagen Type
I(M-60) (Santa Cruz, sc30136), 1: 1:200, 2nd : 1:10,000; Collagen 1A2(M80) (Santa Cruz,
sc28654), 1: 1:200, 2nd : 1:20,000; Collagen Type III(H300) (Santa Cruz, sc28888), 1: 1:200,
2nd : 1:20,000; Tropoelatsin (Elastin Product Co, Inc, PR385), 1: 1:500, 2nd : 1:50,000;
GAPDH (FL-335) (Santa Cruz, sc25778), 1: 1:500, 2nd : 1:20,000; and Alpha SMA, (Abcam,
ab5694), 1: 1:100, 2nd : 1:10,000. All primary antibodies were rabbit IgG. The secondary
antibodies for all were goat anti-rabbit IgG –HRP cat# 31460 from Pierce and developed with
Super signal West Pico chemiluminescent substrate Cat # 34080 from Pierce.
Lung tissue was homogenized with a hand held polytron in buffer contain (20mM Tris-HCl,
pH7.5, 1mM DTT, 1mMEDTA 50mM PMSF plus Protease inhibitor cocktail tablet from
ROCHE) and spun for 10 min at 13000 g to collect total protein. 45 μg of lung total protein
was added 1:2 ratio to Laemmli sample buffer (BioRad-161-0737) containing βME (50 μL
βME in 950 μL sample buffer) then boiled for 3 min. The protein was separated by 10% Tris-
HCl SDS PAGE gel (Bio Rad, 161-1155), then transferred over night at 4°C to a PVDF
Membrane (Perkin Elmer, 370790.) The membrane was blocked with 5% non fat milk in TBS
for 1 hr, then was incubated for 1 hr with primary antibody (antibody in TBS with 2% milk)
After washing 3 times in TBS with 0.5% Tween 20 for 5 min each the membranes were
incubated with secondary goat anti-rabbit IgG –HRP conjugate. After performing the washing
step, the membrane was developed for 5 min with the chemiluminescent substrate (SuperSignal
West Pico kit, 34080, Pierce) and exposed to clear blue X-ray film (Pierce, 34093). Suitable
signal strength was obtained after 2 min exposure.
The same membrane was then stripped for 15 min at room temperature with Restore Western
Blot Stripping Buffer (Pierce, 21059) and then re-blotting following the same above procedures
for internal standard using GAPDH. Assuming equal loading, arsenic did not affect the level
of expression of GAPDH.
Pulmonary Function
Air way responsiveness was measure on live unrestrained mice at various ages, beginning at
28 days after birth. Lung functions were analyzed by comparing the airways responsiveness
to methacholine in unrestrained conscious mice using a Biosystem XA whole body
plethysmographs from Buxco Electronics Inc. (Wilmington, North Carolina) as described
(Hamelmann et al., 1997). The system was calibrated with 1 ml of air for each chamber prior
to use. Mice were then placed in the chamber for 5 to 10 min to allow them to become familiar
with the box and until all breathing was normal and Penh values (enhance pause) were constant.
Enhanced pause (Penh) index of airway hyper-reactivity was used as an indicator of changes
in airway resistance. Penh was calculated by Biosystem XA software using the following
formula: (peak expiratory pressure/peak inspiratory pressure) × (expiratory time - relaxation
time)/relaxation time. Baseline Penh readings were taken and averaged for 5 minutes before
the PBS exposure. Mice were then exposed for 2 min to the nebulized PBS or nebulized
methacholine (1- 100 mg/ml in PBS) (Sigma, A2251) and Penh was recorded for 5 minutes
and averaged. The aerosol delivery system was set to deliver 75 μl of PBS or methacholine per
chamber over the period of 2 min.
In addition to in vivo testing of airway reactivity, pulmonary resistance and compliance were
also tested in additional mice (Robledo et al, 2000). Mice were anesthetized with an
intramuscular injection mixture of ketamine HCL (80 mg/kg), xylaxine (10 mg/kg) and
acepromazine maleate (3 mg/kg). Following this a tracheostomy was performed, inserting a
Teflon IV catheter (20 gauge, Critikon, Tampa Bay, FL) as an endotracheal tube. The mice
were placed on a small animal ventilator (Kent Scientific, Litchfield, CT) under pressure
Lantz et al. Page 5
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
controlled ventilation. Airflow was measured with a pneumotachograph (Fleish #0000,
Instrumentation Associates, New York, NY), which was connected to a differential pressure
transducer (Validyne, Northridge, CA). Pulmonary function measurements were obtained
using a computerized pulmonary function system (PEDS-LAB, Medical Associates Services,
Hatfield, PA) which records airflow and pressure signals, and normalizes them to animal body
weight.
Data Analysis
Statistical analyses were performed using ANOVA (Winer et al., 1991), requiring p<0.05 for
statistical significance.
Results
Twenty eight day old mice exposed to arsenic in utero from conception through birth and
weaning and continuously after weaning, were analyzed for their response to methacholine
challenge. Figure 1A shows the response as a function of baseline responsiveness. As can be
seen, animals that had been exposed to arsenic had increased Penh levels at lower levels of
methacholine challenge. Penh levels were significantly increased at 10 mg/ml methacholine
exposure in animals that had been exposed to 100 ppb arsenic during in utero and postnatal
development. At 33 mg/ml, Penh continued to increase in animals exposed to 100 ppb, while
those exposed to 10 and 50 ppb arsenic also demonstrated a dose-dependent increase in Penh.
To determine if the increases in Penh caused by arsenic exposures was reversible, 28 day old
animals that had been continuously exposed to 50 ppb arsenic since conception were split into
two groups. One group was continued on 50 ppb in their drinking water (50-50 ppb) while
arsenic was eliminated from the drinking water in the second group (50-0 ppb). A parallel
control group was also analyzed. Methacholine-induced changes in Penh were again evaluated
when the animals reached 3 months of age (Figure 1B). As can be seen, removal of the animals
from the arsenic exposure on day 28 did not reduce the increased response to methacholine
challenge. Therefore, alterations in the response to methacholine that were caused by in
utero and early postnatal exposure to arsenic were not reversible by removal of arsenic after
the early developmental time period.
In order to evaluate whether alterations in response to methacholine challenge was specific to
exposure during developmental times, adult male mice were exposed to arsenic in their drinking
water for three months and evaluated for methacholine-induced changes in Penh. Parallel
control animals were also analyzed. Adult only exposure to 100 ppb arsenite in drinking water
did not lead to altered pulmonary response to methacholine challenge (Figure 1C). Therefore,
the alterations in lung function were specific to in utero and early postnatal exposures.
Results from unanaesthetized animals were validated by determining alterations in pulmonary
resistance and compliance in anesthetized animals. As shown in Figure 2, exposure to arsenic
resulted in a dose dependent increase in pulmonary resistance in 4 week old animals. (Figure
2A). This increase was no altered by removal of arsenic from the animals at 28 days of age
(Figure 2B). There were no changes in lung compliance.
Since functional alterations are not affected by removal of the arsenic, we investigated if arsenic
exposure was resulting in airway anatomical structural changes. Changes in airway diameters
and/or airway smooth muscle could account in part for the functional alterations. As a first
measure of airway size, we evaluated the volume density of the airways (percentage of lung
occupied by airways). There was no differences (data not shown), indicting no gross changes
in airway development. Examination of airways from 28 day old animals that had been
immunostained with smooth muscle actin (SMA) antibodies, however, did show an apparent
Lantz et al. Page 6
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
increase in smooth muscle around airways of animals that had been exposed to arsenic (Figure
3).
In order to determine whether quantifiable changes in SMA had occurred we determined the
levels of mRNA and protein as a function of postnatal age (Figure 4). As has been previously
shown (http://lungtranscriptome.bwh.harvard.edu/), mRNA expression of SMA peaked on
postnatal day 7 and subsequently decreased at 12 and 28 days after birth. Exposure to arsenic
during in utero and early postnatal development altered this expression pattern. High levels on
day 7 remained elevated on postnatal days 12 and 28. Whole lung levels of protein expression
followed a similar pattern, that is, elevation of day 7 with subsequent decreases in the control
animals. SMA protein levels remained elevated in arsenic exposed pups.
We have previously shown in adults that chronic exposure to arsenic can lead to a significant
reduction in expression of collagens and elastin in the lung (Lantz and Hays, 2006). Since these
matrix molecules are important for appropriate lung development and since they can also
control smooth muscle proliferation, we examined arsenic induced alterations in expression of
Col1a1, Col1a2, Col3a1 and elastin. In utero and postnatal arsenic exposure resulted in dose
and time dependent alterations in expression of Col1a2, Col3a1 and elastin (Figure 5). The
expression patterns in animals that did not receive arsenic were similar to those that have
previously been reported (http://lungtranscriptome.bwh.harvard.edu/). Rather than
suppression of expression of these matrix genes, arsenic exposure resulted in increases in
expression. Exposure to either 50 or 100 ppb arsenic resulted in increased Col1a2 expression
on postnatal day 7 (Figure 5A). Col3a1 expression was increased on postnatal day 12 by 100
ppb (Figure 5B). The pattern of elastin expression was more complex but, in general, resulted
in increased elastin expression at early postnatal times (Figure 5C). Levels of expression in
arsenic exposed animals were decreased on day 28, similar to what we have previously seen
in adults. Arsenic did not alter the expression pattern of Col1a1 during the early postnatal
periods (data not shown).
While levels of gene expression were altered by arsenic, whole lung levels of matrix protein
were only marginally altered. Analysis of Western blot intensities, normalized to GAPDH
expression levels, showed increased protein expression of Col1a2 on day 12 (control = 1.89 ±
0.09; 50 ppb arsenic = 2.18 ± 0.07; 100 ppb arsenic = 2.56 ± 0.48), decreased protein expression
of Col3a1 on day 12 (control = 1.86 ± 0.09; 100 ppb arsenic = 1.26 ± 0.09) and decreased
protein expression of elastin on day 7 (control = 2.20 ± 0.41; 50 ppb arsenic = 1.47 ± 0.58; 100
ppb arsenic = 0.61 ± 0.35). While these values show trends, only the arsenic-induced decrease
in Col3a1 on day 12 reached significance. Whole lung protein levels for Col1a1, 1a2, 3a1 and
elastin from arsenic exposed animals on day 28 were not different from controls (data not
shown).
Whole lung levels of expression are useful for determination of global expression changes.
However, morphological targeting of analysis can better reflect region differences in
expression. We therefore determined the levels of SMA protein expression around airways.
Values were determined using image analysis as outlined in the methods. Figure 6A shows the
results. Data are presented as total measurements of all airways. Data are also subdivided into
data from airways with diameters less than or greater than 100 μm. When all airways are
combined for analysis, there is an arsenic-induced dose dependent increase in the amount of
SMA around airways of 28 day old mice. A breakdown of the data based on airway diameter
indicates that the majority of this change is due to increased levels around airways that are less
then 100 μm in diameter.
Since we have seen arsenic-induced alterations in SMA expression around airways, we also
examined the levels of collagen around airways as well. Sections were stained with sirius red
Lantz et al. Page 7
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
and levels of collagen were determined by image analysis as outlined in the methods. This stain
does not differentiate collagen subtypes so data reflect total collagen around airways. Data
were also subdivided based on airway diameters. As can be seen in Figure 6B, arsenic exposures
resulted in reduced levels of collagen expression around airways. The suppression of collagen
expression was more evident at 50 ppb exposure levels. In addition, the relative decrease in
collagen expression did not appear to be related to airway diameters, 50 ppb arsenic resulted
in 44,5% decrease in airways with diameters < 100 μm and a 53.1% decrease in airways with
diameters > 100 μm.
Discussion
Using environmentally relevant levels of exposure to arsenic in drinking water, we have shown
that continuous in utero and early postnatal exposure results in both functional and structural
alterations in the lung. Arsenic exposure resulted in increased bronchoconstriction following
methacholine challenge. This alteration in responsiveness could not be reversed by removal
from arsenic exposure. In addition, the alterations in function were the result of exposure during
in utero and early postnatal lung development. Exposure to adults only did not result in the
changes. Associated with these functional changes were alterations in lung structure. Airway
smooth muscle content was increased with increasing arsenic exposure. These changes were
most prominent in airways less than 100 μm in diameter. In addition, arsenic exposure also
altered the expression patterns of several matrix genes that can regulate smooth muscle
proliferation.
Exposure to toxicants during sensitive times of development can produce adverse health
outcomes. For arsenic, this has been demonstrated by Smith et al (2006), who have studied a
human population from Chile that received in utero and early postnatal exposures to high
environmental levels (800 – 900 ppb) of arsenic. These developmental exposures greatly
increased the incidence of lung disease associated with mortalities later in life. Standard
mortality ratios (SMR) from lung cancers, bronchiectasis and other chronic lung diseases were
increased five to ten fold with early postnatal exposure alone. Combined in utero and postnatal
exposures increased the SMR even higher. Our data reported here using 50 to 100 ppb arsenic
exposures are therefore relevant for providing potential mechanisms that can lead to human
diseases.
A significant proportion of adult lung disease originates in utero or early infancy (Merkus,
2003). A number of agents have been shown to affect normal lung growth and development
when administered in utero or in the first month after birth. Maternal smoking has been
associated with decreased elastic tissue and increased alveolar size (Collins et al, 1985). For
postnatal exposures, the more extensively studied compounds are dexamethasone, hyperoxia
and Sendai viral infections. Dexamethasone, administered in a critical time period between 4
and 14 days after birth, resulted in larger alveoli on day 14. This enlargement was still present
on day 60 after birth (Blanco et al, 1989). Dexamethasone had no effect when given after day
14 (Blanco and Frank, 1993). Similarly, the postnatal age at the onset of hyperoxic exposure
was found to affect the expression of tropoelastin (Bruce et al, 1996). Exposure from 3 to 13
days of age interfered with the normal increase in tropoelastin expression seen during this time.
Hyperoxic exposure in weeks 3 and 4 was still capable of increasing alveolar size and
decreasing alveolar number (Blanco and Frank, 1993). In these experiments, decreases in
septation were associated with decreases in elastin fiber length and enlarged alveoli.
A similar sensitive period was noted in rats that had been infected with Sendai virus (Castleman
et al, 1988). Exposure of 5-day-old rats produced more severe changes in structure and function
than inoculations on day 25. In these studies, lungs from virus exposed rats appeared to have
mild alveolar emphysema. Lung function revealed elevated resistance and decreased
Lantz et al. Page 8
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
compliance, and increased airway hyperresponsiveness (Sorkness et al, 1991). These
alterations were not compensated for when rats were tested at 39 days after inoculation.
In addition to alterations caused by postnatal exposure to toxicants and infectious agents,
prenatal exposure to cigarette smoke results in increased airway hyperreactivity (AHR) (Singh
et al, 2003). The alterations in lung function occurred only in animals that were exposed in
utero. Postnatal exposures or exposures in adults did not lead to increases in AHR.
In the current report, we have provided continual in utero and postnatal exposures similar to
what would be experienced in the real world. As such we can not differentiate between the
effects of in utero or postnatal alone. Defining the sensitive exposure window will be important
in future work. Our results however, are similar to other agents that act either in utero or
postnatally. Alterations in function are caused by exposures during these developmental times
and are irreversible even with withdrawal of the arsenic.
Effects of agents that lead to structural and functional alterations on gene expression have only
been examined in a limited number of studies in neonatal lung. Hyperoxia in neonates has been
shown to induce antioxidant enzymes (Clerch and Massaro, 1992) and surfactant protein A
(D'Angio et al, 1997) while down-regulating tropoelastin expression (Bruce, 1991). Treatment
of neonatal lung with dexamethasone has also been shown to down-regulate cellular retinol
binding protein 1 (Whitney et al, 1999).
We have previously shown down regulation of extracellular matrix gene expression in the lung
following chronic exposure in adult mice (Lantz and Hays, 2006). Based on the importance of
these proteins in lung development we tested whether arsenic could down regulate collagen
and elastin expression during postnatal lung development. However, rather than inhibiting the
expression, arsenic enhanced the levels of whole lung gene expression for Col1a2 and Col3a1
in a time dependent manner. This suggests that arsenic is interacting with the normal
developmental process to alter expression of these matrix genes. Whole lung protein levels
however were not significantly altered. One explanation is that mRNA levels of expression
may be increasing to compensate for losses of proteins, so that normal levels of matrix protein
required for developmental processes are maintained. We have previously demonstrated that
arsenic can induce increased expression of matrix metalloproteinase-9 (MMP-9) (Olsen et al,
2008). Similar arsenic-induced changes in MMP-9 during development would degrade matrix.
Increases in mRNA expression could be a compensatory response.
While whole lung levels of matrix proteins were unchanged, regional decreases in total collagen
in adventia around airways was seen in 28 day old mice exposed to arsenic during development.
This localized regional decrease in collagen could contribute to the increased smooth muscle
around airways (Dekkers et al, 2007; Parameswaran et al, 2006), similar to collagen knockout
mice. Elastin and collagen knockout mice both show hyperproliferation of smooth muscle
around blood vessels (Karnik et al, 2003, Liu et al, 1997). Therefore decreased expression of
collagen around airways may also contribute to the increased levels of smooth muscle.
Increases in smooth muscle are associated with increases in airway hyperresponsiveness
(Martin et al, 2000; Cockcroft and Davis, 2006). Alterations in smooth muscle have best been
characterized in a primate model (Plopper et al, 2007). Exposure to ozone during postnatal
lung development led to alterations in airway smooth muscle orientation (Fanucchi et al,
2006). However, there was no change in the thickness or abundance of smooth muscle around
the airways following ozone only exposures. Alterations of function were only noted during a
subsequent allergen challenge, with no changes in baseline lung function (Tran et al, 2004).
Exposure to house dust mite allergen in the same developmental primate model did result in
increased abundance and mass of smooth muscle around airways. Rodents sensitized with
ovalbumin also show increased airway hyper-responsiveness to challenge, which is associated
Lantz et al. Page 9
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
with increased airway smooth muscle (Moir et al, 2003). Arsenic-induced increases in smooth
muscle mass, particularly around small airways is therefore more like alterations seen with
allergen senitization.
Alterations in collagen expression can not be the only factor leading to increased smooth muscle
mass. Smooth muscle alterations were greatest in the smaller airways, while decreases in
collagen did not show an airway size preference. While these results are not as we expected
from our adult exposures, they do demonstrate that arsenic is interacting with the expression
of important developmental genes, which contributes to alterations in lung structure and
function.
There is concern about arsenic levels in laboratory diets affecting and masking results obtained
with animals exposed to low dose arsenic in drinking water (Kozul et al, 2008). Differences
in gene expression were evident when diets containing 400 ppb arsenic were compared with
diets that had low levels of arsenic (20 ppb). Our diets contained 40 ppb total arsenic, which
is higher than the AIN-76A diet but an order of magnitude lower than the LRD-5001 diet
studied by Kozul. The diet we selected was based on the ability to alter nutritional items for
future studies. Based on the NHEXAS data (Pellizzari and Clayton, 2006), the average human
adult arsenic exposure levels are around 20 ppb. Children's exposure levels were about twice
as high. The levels found in our diets were close to the human exposure levels. It is difficult
to calculate the daily intake of arsenic during development, due to multiple sources of exposure
(i.e. in utero, nursing after birth, weaned pups). However, assuming total bioavailablity from
both water and food, an assumption that is probably an over estimation of the actual exposure
levels, we would expect the arsenic food levels to raise the total arsenic exposure by around
20 ppb. This would elevate the total arsenic intake by about 25% and 15% for 50 and 100 ppb
water, respectively. However, since both the food and water intake levels are similar to those
seen in real world situations, we feel that our data still represent arsenic-induced changes that
are seen at environmentally relevant levels.
It is interesting to speculate on which developmental pathways may be targeted by the arsenic
exposures. The results from our current experiments show altered matrix expression and
increased levels of smooth muscle around airways and increases in smooth muscle actin in
whole lung. Our previous research has shown that arsenic administered at levels similar to
those used in these experiments, increased MMP-9 expression and altered wound repair
processes (Olsen et al, 2008). One growth factor that plays a role in all of these processes is
TGF-β (Xie et al, 2007; Ohbayashi and Shimokata, 2005). Arsenic-induced increases in TGF-
β1 have been reported for both acute and chronic exposures to arsenite. Injection of arsenite
resulted in increased levels of TGF- β1 in kidney (Kimura et al, 2006). Long term ingestion of
200 ppm arsenite for 10 months increased TGF- β1 gene expression in the liver (Wu et al,
2008). In this case, gene expression for procollagen 1 and 3 and for smooth muscle actin were
also increased. Therefore, arsenic-induced changes in TGF- β1 signaling may contribute to the
alterations during development. We have previously identified the β-catenin pathway as an
important target of arsenic in the lung during in utero development (Petrick et al, 2008). Wnt
and β-catenin signaling, which can be modulated by TGF-β (Caraci et al, 2008), can also lead
to increases in airway smooth muscle (Nunes et al, 2008). The effects of arsenic on TGF-β
expression during development are currently under investigation in our laboratory.
We have shown continuous in utero and early postnatal exposure to arsenic at environmentally
relevant levels results in irreversible functional and structural alterations in the lung. These
results demonstrate the importance of exposures during sensitive developmental time points
and suggest that interventions must also occur during these developmental time points in order
to be effective.
Lantz et al. Page 10
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Acknowledgements
This work was supported in part by the Superfund Basic Research Program NIEHS Grant Number P42-ES-04940 and
the Southwest Environmental Health Sciences Center P30-ES-06694.
References
Blanco LN, Frank L. The formation of alveoli in rat lung during the third and fourth postnatal weeks:
effect of hyperoxia, dexamethasone, and deferoxamine. Ped Res 1993;34:334–340.
Blanco LN, Massaro GD, Massaro D. Alveolar dimensions and number, developmental and hormonal
regulation. Am J Physiol 1989;257:L240–L247. [PubMed: 2801952]
Bruce MC. Developmental changes in tropoelastin mRNA levels in rat lung: evaluation by in situ
hybridization. Amer J Respir Cell Mol Biol 1991;5:344–350. [PubMed: 1910819]
Bruce MC, Honaker C, Karathanasis P. Postnatal age at onset of hyperoxic exposure influences
developmentally regulated tropoelastin gene expression in the neonatal rat lung. Am J Respir Cell Mol
Biology 1996;14:177–185.
Camateros P, Tamaoka M, Hassan M, Marino R, Moisan J, Marion D, Guiot MC, Martin JG. Chronic
asthma-induced airway remodeling is prevented by toll-like receptor-7/8 ligand S28463. Am J Respir
Crit Care Med 2007;175:1241–1249. [PubMed: 17400732]
Caraci F, Gili E, Calafiore M, Failla M, La Rosa C, Crimi N, Sortino MA, Nicoletti F, Copani A, Vancheri
C. TGF-beta1 targets the GSK-3beta/beta-catenin pathway via ERK activation in the transition of
human lung fibroblasts into myofibroblasts. Pharmacol Res 2008;57:274–282. [PubMed: 18346908]
Castleman WL, Sorkness RL, Lemanske RF, Grasee G, Suyemoto MM. Neonatal viral bronchiolitis and
pneumonia induces bronchiolar hypoplasia and alveolar dysplasia in rats. Lab Invest 1988;59:387–
396. [PubMed: 3411939]
Chen CJ, Kuo TL, Wu MM. Arsenic and cancers (letter). Lancet 1988;1988:414–415. [PubMed:
2893213]
Chiou HY, Hsueh YM, Liaw KF, Horng SF, Chiang MH, Pu YS, Lin JS, Huang CH, Chen CJ. Incidence
of internal cancers and ingested inorganic arsenic: a seven-year follow-up study in Taiwan. Cancer
Res 1995;55:1296–1300. [PubMed: 7882325]
Clerch LB, Massaro D. Rat lung antioxidant enzymes: differences in perinatal gene expression and
regulation. Am J Physiol 1992;263:L466–L470. [PubMed: 1415724]
Cockcroft DW, Davis BE. Mechanisms of airway hyperresponsiveness. J Allergy Clin Immunol
2006;118:551–559. [PubMed: 16950269]
Collins MH, Moessinger AC, Kleinerman J, Bassi J, Rosso P, Collins AM, James S, Blanc WA. Fetal
lung hypoplasia associated with maternal smoking: a morphometric analysis. Ped Res 1985;19:408–
412.
Concha G, Vogler G, Lezcano D, Nermell B, Vahter M. Exposure to inorganic arsenic metabolites during
early human development. Toxicol Sci 1998;44:185–190. [PubMed: 9742656]
D'Angio CT, Finkelstein JN, Lomonaco MB, Paxhia A, Wright SA, Baggs RB, Notter RH, Ryan RM.
Changes in surfactant protein gene expression in a neonatal rabbit model of hyperoxia-induced
fibrosis. Am J Physiol 1997;272:L720–L730. [PubMed: 9142947]
Dekkers BGJ, Schaafsma D, Nelemans SA, Zaagsma J, Meurs H. Extracellular matrix proteins
differentially regulate airway smooth muscle phenotype and function. Am J Physiol Lung Cell Mol
Physiol 2007;292:L1405–L1413. [PubMed: 17293376]
Fanucchi MV, Plopper CG, Evans MJ, Hyde DM, Van Winkle LS, Gershwin LJ, Schelegle ES. Cyclic
exposure to ozone alters distal airway development in infant rhesus monkeys. Am J Physiol Lung
Cell Mol Physiol 2006;291:L644–L650. [PubMed: 16648242]
Hamelmann E, Schwarze J, Takeda K, Oshiba A, Larsen GL, Irvin CG, Gelfand EW. Noninvasive
measurement of airway responsiveness in allergic mice using barometric plethysmography. Am J
Respir Crit Care Med 1997;156:766–775. [PubMed: 9309991]
Hopenhayn-Rich C, Biggs ML, Smith AH. Lung and kidney cancer mortality associated with arsenic in
drinking water in Cordoba, Argentina. Int J Epidemiol 1998;27:561–569. [PubMed: 9758107]
Lantz et al. Page 11
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Hopenhayn-Rich C, Browning SR, Hertz-Picciotto I, Ferreccio C, Gibb H. Chronic arsenic exposure and
risk of infant mortality in two areas of Chile. Environ Health Perspect 2000;108:667–673. [PubMed:
10903622]
Hopenhayn C, Ferreccio C, Browning SR, Huang B, Peralta C, Gibb H, Hertz-Picciotto I. Arsenic
exposure from drinking water and birth weight. Epidemiol 2003;14:593–602.
Karnik SK, Brooke BS, Bayes-Genis A, Sorensen L, Wythe JD, Schwartz RS, Keating MT, Li DY.
Development and disease: a critical role for elastin signaling in vascular morphogenesis and disease.
Develop 2003;130:411–423.
Kimura A, Ishida Y, Hayashi T, Wada T, Yokoyama H, Sugaya T, Mukaida N, Kondo T. Interferon-
gamma plays protective roles in sodium arsenite-induced renal injury by up-regulating intrarenal
multidrug resistance-associated protein 1 expression. Am J Pathol 2006;169:1118–1128. [PubMed:
17003472]
Kozul CD, Nomikos AP, Hampton TH, Warnke LA, Gosse JA, Davey JC, Thorpe JE, Jackson BP, Ihnat
MA, Hamilton JW. Laboratory diet profoundly alters gene expression and confounds genomic
analysis in mouse liver and lung. Chem Biol Interact 2008;173:129–140. [PubMed: 18396267]
Lantz RC, Hays AM. Role of oxidative stress in arsenic-induced toxicity. Drug Metab Rev 2006;38:791–
804. [PubMed: 17145702]
Last JA, Ward R, Temple L, Pinkerton KE, Kenyon NJ. Ovalbumin-induced airway inflammation and
fibrosis in mice also exposed to ultrafine particles. Inhal Toxicol 2004;16:93–102. [PubMed:
15204782]
Liu J, Xie Y, Ducharme MK, Shen J, Tennant R, Diwan BA, Waalkes MP. Global gene expression
associated with hepatocarcinogenesis in adult male mice induced by in utero arsenic exposure.
Environ Health Perspect 2006;114:404–411. [PubMed: 16507464]
Liu X, Wu H, Byrne M, Krane S, Jaenisch R. Type III collagen is critical for collagen I fibrillogenesis
and for normal cardiovascular development. Proc Natl Acad Sci 1997;94:1852–1856. [PubMed:
9050868]
Mariani TJ, Reed JJ, Shapiro SD. Expression profiling of the developing mouse lung: insights into the
establishment of the extracellular matrix. Am J Respir Cell Mol Biol 2002;26:541–548. [PubMed:
11970905]
Martin JG, Duguet A, Eidelman DH. The contribution of airway smooth muscle to airway narrowing and
airway hyperresponsiveness in disease. Eur Respir J 2000;16:349–354. [PubMed: 10968513]
Mazumder DN. Effect of drinking arsenic contaminated water in children. Indian Pediatr 2007;44:925–
927. [PubMed: 18175848]
Mazumder DN, Haque R, Ghosh N, De BK, Santra A, Chakraborti D, Smith AH. Arsenic in drinking
water and the prevalence of respiratory effects in West Bengal, India. Int J Epidemiol 2000;29:1047–
1052. [PubMed: 11101546]
Merkus PJ. Effects of childhood respiratory disease on the anatomical and functional development of the
respiratory system. Paed Respir Reviews 2003;4:28–39.
Moir LM, Leung SY, Eynott PR, McVicker CG, Ward JP, Chung KF, Hirst SJ. Repeated allergen
inhalation induces phenotypic modulation of smooth muscle in bronchioles of sensitized rats. Am J
Physiol Lung Cell Mol Physiol 2003;284:L148–L159. [PubMed: 12388362]
Nunes RO, Schmidt M, Dueck G, Baarsma H, Halayko AJ, Kerstjens HA, Meurs H, Gosens R. GSK-3/
beta-catenin signaling axis in airway smooth muscle: role in mitogenic signaling. Am J Physiol Lung
Cell Mol Physiol 2008;294:L1110–L1118. [PubMed: 18390827]
Ohbayashi H, Shimokata K. Matrix metalloproteinase-9 and airway remodeling in asthma. Current Drug
Targets - Inflammation Allergy 2005;4:177–181. [PubMed: 15853739]
Olsen C, Liguori A, Zong Y, Lantz RC, Burgess J, Boitano S. Arsenic upregulates MMP-9 and inhibits
wound repair in human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2008;295:L293–
L302. [PubMed: 18539681]
Parameswaran K, Willems-Widyastuti A, Alagappan VKT, Radford K, Kranenburg AR, Sharma HS.
Role of extracellular matrix and its regulators in human airway smooth muscle biology. Cell Biochem
Biophys 2006;44:139–146. [PubMed: 16456242]
Lantz et al. Page 12
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Pellizzari ED, Clayton CA. Assessing the measurement precision of various arsenic forms and arsenic
exposure in the National Human Exposure Assessment Survey (NHEXAS), Environ. Health Perspect
2006;114:220–227.
Petrick JS, Blachere FM, Selmin O, Lantz RC. Inorganic arsenic as a developmental toxicant: in utero
exposure and alterations in developing lung. Mol Nut Food Res. 2008in press
Plopper CG, Smiley-Jewell SM, Miller LA, Fanucchi MV, Evans MJ, Buckpitt AR, Avdalovic M,
Gershwin LJ, Joad JP, Kajekar R, Larson S, Pinkerton KE, Van Winkle LS, Schelegle ES, Pieczarka
EM, Wu R, Hyde DM. Asthma/allergic airways disease: does postnatal exposure to environmental
toxicants promote airway pathobiology? Toxicol Pathol 2007;35:97–110. [PubMed: 17325978]
Robledo RF, Young RS, Lantz RC, Witten ML. Short-term pulmonary response to inhaled JP-8 jet fuel
aerosol in mice. Toxicol Pathol 2000;28:656–63. [PubMed: 11026600]
Rosenberg HG. Systemic arterial disease and chronic arsenicism in infants. Arch Pathol 1974;97:360–
365. [PubMed: 4825098]
Shen J, Liu J, Xie Y, Diwan BA, Waalkes MP. Fetal onset of aberrant gene expression relevant to
pulmonary carcinogenesis in lung adenocarcinoma development induced by in utero arsenic
exposure. Toxicol Sci 2007;95:313–320. [PubMed: 17077188]
Singh SP, Barrett EG, Kalra R, Razani-Boroujerdi S, Langley RJ, Kurup V, Tesfaigzi Y, Sopori ML.
Prenatal cigarette smoke decreases lung cAMP and increases airway hyperresponsiveness. Am J
Respir Crit Care Med 2003;168:342–347. [PubMed: 12791581]
Smith AH, Goycolea M, Haque R, Biggs ML. Marked changes in bladder and lung cancer mortality in
a region of northern Chile due to arsenic in drinking water. Am J Epidemiol 1998;147:660–669.
[PubMed: 9554605]
Smith AH, Marshall G, Yuan Y, Ferreccio C, Liaw J, von Ehrenstein O, Steinmaus C, Bates MN, Selvin
S. Increased mortality from lung cancer and bronchiectasis in young adults after exposure to arsenic
in utero and in early childhood. Environ Health Perspect 2006;114:1293–1296. [PubMed: 16882542]
Sorkness R, Lemanske RF Jr, Castleman WL. Persistent airway hyperresponsiveness after neonatal
bronchiolitis in rats. J Appl Physiol 1991;70:375–383. [PubMed: 1849132]
Tran MU, Weir AJ, Fanucchi MV, Rodriguez AE, Pantle LM, Smiley-Jewell SM, Van Winkle LS, Evans
MJ, Miller LA, Schelegle ES, Gershwin LJ, Hyde DM, Plopper CG. Smooth muscle hypertrophy in
distal airways of sensitized infant rhesus monkeys exposed to house dust mite allergen. Clin Exp
Allergy 2004;34:1627–1633. [PubMed: 15479280]
Vahter M. Health effects of early life exposure to arsenic. Basic Clin Pharmacol Toxicol 2008;102:204–
211. [PubMed: 18226075]
von Ehrenstein OS, Mazumder DN, Yuan Y, Samanta S, Balmes J, Sil A, Ghosh N, Hira-Smith M, Haque
R, Purushothamam R, Lahiri S, Das S, Smith AH. Decrements in lung function related to arsenic in
drinking water in West Bengal, India. Am J Epidemiol 2005;162:533–541. [PubMed: 16093295]
Waalkes MP, Ward JM, Liu J, Diwan BA. Transplacental carcinogenicity of inorganic arsenic in the
drinking water: induction of hepatic, ovarian, pulmonary, and adrenal tumors in mice, Toxicol. Appl
Pharmacol 2003;186:7–17.
Wei S, Bellusci S, Warburton D. Lung development and adult lung diseases. Chest 2007;132:651–656.
[PubMed: 17699136]
Weibel, ER. Stereological Methods Vol 1: Practical Methods for Biological Morphometry. Academic
Press; London: 1979.
Whitney D, Massaro GD, Massaro D, Clerch LB. Gene expression of cellular retinoid-binding proteins:
modulation by retinoic acid and dexamethasone in postnatal rat lung. Ped Res 1999;45:2–7.
Winer, BJ.; Brown, DR.; Michels, KM. Statistical Principles in Experimental Design. McGraw-Hill; New
York: 1991.
Wlodarczyk B, Bennett GD, Calvin JA, Craig JC, Finnell RH. Arsenic-induced alterations in embryonic
transcription factor gene expression: implications for abnormal neural development. Dev Genet
1996;18:306–315. [PubMed: 8754282]
Wu J, Liu J, Waalkes MP, Cheng ML, Li L, Li CX, Yang Q. High dietary fat exacerbates arsenic-induced
liver fibrosis in mice. Exp Biol Med 2008;233:377–384.
Lantz et al. Page 13
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Wu MM, Kuo TL, Hwang YH, Chen CJ. Dose–response relation between arsenic concentration in well
water and mortality from cancers and vascular disease. Am J Epidemiol 1989;130:1123–1132.
[PubMed: 2589305]
Xie S, Sukkar MB, Issa R, Khorasani NM, Chung KF. Mechanisms of induction of airway smooth muscle
hyperplasia by transforming growth factor-β. Am J Physiol Lung Cell Mol Physiol 2007;293:L245–
L253. [PubMed: 17468136]
Lantz et al. Page 14
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Response to methacholine challenge in unrestrained conscious mice
A - Response to methacholine challenge in 28 day old mice that had been continuously exposed
to arsenic in utero and during postnatal development. Animals that had been exposed to higher
levels of arsenic during development responded with increased Penh levels at similar
methacholine challenge levels. Values are means ± sem compared to control animals. N=3 in
at each arsenic dose. *=significantly different from control (p<0.05).
B – Response of twelve week old mice to methacholine challenge. All arsenic exposed mice
were exposed to 50 ppb arsenic during in utero and early postnatal development. At 28 days
of age, animals were split into two groups. One group continued to receive 50 ppb arsenic in
their drinking water (50-50 ppb) while arsenic was removed from the water given to the second
Lantz et al. Page 15
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
group (50-0 ppb). Controls received no arsenic throughout. Animals were kept on this regimen
until they reached three months of age, when they were tested for their response to methacholine
challenge. Removal of arsenic from the water at 28 days of age did not return methacholine
response to control levels. The response of the 50-50 and 50-0 animals were not significantly
different from each other. Both 50-50 animals and 50-0 animals showed increased response to
methacholine compared to controls. (controls, N=5; 50-50, N=4; 50-0, N=3). Values are mean
± sem. *=significantly different from control (p<0.05).
C- Response to methacholine challenge in mice that have only been exposed to arsenic as
adults. Adult mice were given 100 ppb arsenic in their drinking water for three months and
tested for alterations in Penh after methacholine challenge. Values for animals exposed to 100
ppb arsenic were not different from animals that received no arsenic in their water. (controls,
N=4; 100 ppb, N=6). Values are mean ± sem.
Lantz et al. Page 16
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2. Pulmonary resistance was measured in anesthetized mice
A – Mice were exposed continuously during in utero and early postnatal development to 50 or
100 ppb arsenic. At 28 days of age, mice were anesthetized and pulmonary resistance was
measured. Arsenic exposure resulted in increased resistance in a dose dependent manner.
(Controls, N=4; 50 ppb, N=4; 100 ppb N=5), Values are mean ± sem. A=significantly different
from Control (p<0.05). B=significantly different from Control and 50 ppb arsenic (p<0.05).
B – Pulmonary resistance in twelve week old mice. All arsenic exposed mice were exposed to
50 ppb arsenic during in utero and early postnatal development. At 28 days of age, animals
were split into two groups. One group continued to receive 50 ppb arsenic in their drinking
water (50-50 ppb) while arsenic was removed from the water given to the second group (50-0
ppb). Controls received no arsenic throughout. Removal of arsenic from the water at 28 days
of age did not return of pulmonary resistance to control levels. Both 50-50 animals and 50-0
animals showed increased resistance compares to controls. (controls, N=4; 50-50, N=7; 50-0,
N=3). Values are mean ± sem. A=significantly different from Control (p<0.05).
Lantz et al. Page 17
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3. Airway smooth muscle
Photomicrograph of smooth muscle actin immunostaining in 28 day old mice. Mice that had
received 100 ppb arsenic continuously during in utero and early postnatal development showed
an apparent increase in the level of smooth muscle actin staining (white staining) around
airways (A). Bar in lower right corner = 100 μm.
Lantz et al. Page 18
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4. Smooth muscle actin mRNA and protein levels from whole lung
A – mRNA levels of SMA as a function of postnatal age are plotted from quantitative RT-
PCR. Arsenic at 50 and 100 ppb led to increased levels of SMA gene expression in the whole
lung. N=3 for each arsenic dose and postnatal age. Values are mean ± sem. A=100 ppb
significantly different from control in 12 day old animals. B=50 and 100 ppb significantly
different from controls in 28 day old animals. (p<0.05)
B – SMA protein levels as a function of postnatal age are plotted from Western blot analysis.
During normal development, protein levels are increased on postnatal day 7 and then decrease
on day 12 and 28. Arsenic increased the levels of SMA expression on day 12. These increases
were maintained on day 28. N=3 for each arsenic dose and postnatal age. A=50 and 100 ppb
significantly different from control in 12 day old animals. B=100 ppb significantly different
from controls in 28 day old animals. (p<0.05).
Lantz et al. Page 19
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 5. Extracellular matrix gene expression
Quantitative RT-PCR for collagen 1a2 (A) and 3a1 (B) and elastin (C) show arsenic-induced
changes in mRNA levels. For Col1a2, arsenic increased the levels of expression of postnatal
day 7, while for col3a1, arsenic increased the levels of expression of postnatal day 12.
Alterations in elastin expression were more complex but in general arsenic increased
expression. On postnatal day 28, levels of expression in arsenic exposed animals were at or
below control levels. Values are mean ± sem. N=3-6 animals/dose/age. Levels of expression
were normalized to GAPDH expression. A=50 and 100 ppb significantly different from Control
in 7 day old animals; B=100 ppb significantly different from Control in 12 day old animals.
(p<0.05).
Lantz et al. Page 20
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 6. Quantitative image analysis of the levels of SMA and collagen staining
A – Quantitative image analysis of the levels of smooth muscle actin staining. The area of
staining around airways was normalized to the area of the airway epithelial basement
membrane. Combining data from all airways shows an arsenic-dependent increase in the levels
of SMA. When the airways are segregated by diameter (< 100 μm and > 100 μm), the majority
of the changes in SMA are seen in the smaller airways. (Control, 12 airways from 4 mice; 50
ppb, 17 airways from 4 mice; 100 ppb, 13 airways from 4 mice). Values are mean ± sem.
A=significantly different from Controls; B=significantly different from Controls and 50 ppb.
(p<0.05).
B - Quantitative image analysis of the levels of collagen around airways. The area of staining
around airways was normalized to the area of the airway epithelial basement membrane.
Combining data from all airways shows an arsenic-dependent increase in the levels of collagen.
The effects of arsenic are independent of the airway diameter. (< 100 μm and > 100 μm),
(Control, 12 airways from 4 mice; 50 ppb, 17 airways from 4 mice; 100 ppb, 13 airways from
4 mice). Values are mean ± sem. A=significantly different from Controls. (p<0.05).
Lantz et al. Page 21
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Lantz et al. Page 22
Table 1
Real Time PCR Primers
Elastin:
Forward: 5-TGG AGG CAA GGG AGC AAG AAA C-3
Reverse: 5-TAA CAG AAC AGA GAG TGC TGT GGG-3
Collagen 1A1:
Forward: 5-GAG CGG AGA GTA CTG GAT CG-3
Reverse: 3-GTT CGG GCT GAT GTA CCA GT-3
Collagen 1A2:
Forward: 5-GGA GGG AAC GGT CCA CGA T-3
Reverse: 5-GAG TCC GCG TAT CCA CAA-3
Collagen 3a1:
Forward: 5-AAG TTC ACC AGC AAC AGC AG-3
Reverse: 5-TTG GTT AGC CAT GTA GAG CG-3
Smooth muscle actin:
Forward: 5-CTT CCA GCC ATC TTT CAT TGG-3
Reverse: 5-ATA TCA CAC TTC ATG ATG CTG TTA TAG GT-3
Beta-Actin:
Forward: 5-AGA GGG AAA TCG TGC GTG AC-3
Reverse: 5-CAA TAG TGA TGA CCT GGC CGT-3
Toxicol Appl Pharmacol. Author manuscript; available in PMC 2010 February 15.
... Our results showed a negative correlation between lung function parameters (FEV1/FVC ratio, FEV1%, FCV%, as well as FEF 25%) and serum arsenic levels among the exposed individuals. Some animal studies have noted the fibrotic lung changes that occur as a result of arsenic exposure (Lantz et al. 2009;Petrick et al. 2009). It is still unclear as to how inhalation of arsenic dust particles over a period of time causes a decline in pulmonary function; however, some animal and human clinical studies have claimed that it may be the result of the arsenic particles depositing within the respiratory epithelium, causing irreversible structural deformities and subsequent functional abnormalities (respiratory function tests) (Gerhardsson et al. 1988). ...
... It is still unclear as to how inhalation of arsenic dust particles over a period of time causes a decline in pulmonary function; however, some animal and human clinical studies have claimed that it may be the result of the arsenic particles depositing within the respiratory epithelium, causing irreversible structural deformities and subsequent functional abnormalities (respiratory function tests) (Gerhardsson et al. 1988). Lantz et al. (2009) reported that the increase in arsenic exposure caused tissue inflammation, as well altered the morphology of lung tissue within animal models. ...
Article
Full-text available
Objective Copper smelter workers are exposed to harmful chemical agents in dust and fumes which contain harmful metals such as copper and arsenic. These substances are known to be respiratory irritants.Methods This study aimed at investigating the effect of occupational exposure to copper and arsenic on the respiratory system. A group of 75 male exposed workers, and 75 male administrative employees (control group) were recruited from a secondary copper smelting factory.Full history, complete clinical examination, ventilatory function parameters (FVC, FEV1, FVC/FEV1 and FEF), and chest X-ray were done for both groups. Serum levels of ICAM-1 and IL8 (as markers of epithelial injury) were measured by ELISA. Serum copper and arsenic were measured by atomic absorption spectrophotometer.ResultsThe exposed group was associated with increased respiratory symptoms, higher serum copper, arsenic, and ICAM-1and Il-8 as compared to the control group. There was a significant decrease in ventilatory parameters among the exposed group: 58.7% of the exposed group had restrictive lung impairment, 40% had obstructive impairment. In the exposed group a positive correlation between serum copper, arsenic and serum ICAM and IL8 was found. While a negative correlation was observed between both serum ICAM, IL8 and ventilatory parameters among the exposed group. Moreover, 36% of the exposed group had radiological infiltrates on chest X.ray.Conclusion Occupational exposure to copper and arsenic was associated with ventilatory and radiological impairment, with a corresponding increase in the serum level of ICAM-1 and IL8, which can be used as biomarkers for pulmonary impairment among copper smelter workers.
... In this context, prenatal exposure of mice to high arsenic concentrations is known to produce cell damaging effects [16,26,27]. For instance, a low dose of 50 ppb arsenic was shown to induce learning impairments [28] and liver dysfunctions in mice [29]. Adverse outcomes of low doses of arsenic on pre-and post-natal development of mice have been reported [30]. ...
... Arsenic has also been associated with increased susceptibility to respiratory infections and cardiovascular disease. Interestingly, fetuses are extremely vulnerable to the effects of heavy metals, particularly arsenic (Lantz et al., 2009;Vahter, 2009;Ahsan et al., 2006;Navas-Acien et al., 2005). ...
Article
Full-text available
Arsenic toxicity is a global health problem affecting millions of people. Contamination is caused by arsenic from natural geological sources leaching into aquifers, contaminating drinking water, and may also be caused by mining and other industrial processes. Acute arsenic poisoning is associated with nausea, vomiting, abdominal pain, and severe diarrhea. Chronic arsenic toxicity results in multisystemic diseases leading to central nervous system (CNS) impairments such as cognitive or intellectual deficits in children. Over the past ten years, arsenic contamination has been reported in northern Thailand. The Ministry of Public Health; Thailand, Forensic Science Institute Tham-masat University, and the Research Center to Promote Safety and Prevent Injuries in Children at the Ramathibodi Hospital compiled a report on the health impact of the population within a 10 kilometer radius around a mine tailing in the Phichit, Phitsanulok, and Phetchabun Provinces of Thailand. It showed that more than 30 % of chil-dren (aged 8–13 years) had higher than normal arsenic contamination levels based on the Agency for Toxic Sub-stances and Disease Registry (ATSDR). After the publication of that report, the mine was temporarily closed in 2016. Based on this data, this research aimed to follow arsenic contamination after the mining operation had stopped operation for three years. The study showed that 4.5 % of school aged children had levels of inorganic arsenic in their urine, higher than the normal range (ATSDR), showing clearly that inorganic arsenic contamination is still above the normal range in children living near an inactive mining site. Therefore, monitoring heavy metal contamination in Thailand and the health effects on vulnerable children who live near mines during regular oper-ation or after being temporarily suspended can prevent and mitigate possible health impacts.
... Arsenic has also been associated with increased susceptibility to respiratory infections and cardiovascular disease. Interestingly, fetuses are extremely vulnerable to the effects of heavy metals, particularly arsenic (Lantz et al., 2009;Vahter, 2009;Ahsan et al., 2006;Navas-Acien et al., 2005). ...
Article
Full-text available
Arsenic toxicity is a global health problem affecting millions of people. Contamination is caused by arsenic from natural geological sources leaching into aquifers, contaminating drinking water, and may also be caused by mining and other industrial processes. Acute arsenic poisoning is associated with nausea, vomiting, abdominal pain, and severe diarrhea. Chronic arsenic toxicity results in multisystemic diseases leading to central nervous system (CNS) impairments such as cognitive or intellectual deficits in children. Over the past ten years, arsenic contamination has been reported in northern Thailand. The Ministry of Public Health; Thailand, Forensic Science Institute Tham-masat University, and the Research Center to Promote Safety and Prevent Injuries in Children at the Ramathibodi Hospital compiled a report on the health impact of the population within a 10 kilometer radius around a mine tailing in the Phichit, Phitsanulok, and Phetchabun Provinces of Thailand. It showed that more than 30 % of children (aged 8-13 years) had higher than normal arsenic contamination levels based on the Agency for Toxic Substances and Disease Registry (ATSDR). After the publication of that report, the mine was temporarily closed in 2016. Based on this data, this research aimed to follow arsenic contamination after the mining operation had stopped operation for three years. The study showed that 4.5 % of school aged children had levels of inorganic arsenic in their urine, higher than the normal range (ATSDR), showing clearly that inorganic arsenic contamination is still above the normal range in children living near an inactive mining site. Therefore, monitoring heavy metal contamination in Thailand and the health effects on vulnerable children who live near mines during regular operation or after being temporarily suspended can prevent and mitigate possible health impacts.
... This suggests that arsenic exposure is likely linked to restrictive lung diseases, rather than obstructive lung diseases like COPD or chronic bronchitis (Sanchez et al. 2018). It is thought that arsenic exposure causes altered wound healing, lung injury, extracellular matrix remodeling, and inflammation which all contributes to pulmonary fibrosis, a restrictive lung disease Fig. 1 Average concentrations of urinary metals (A), urinary arsenic compounds (B), and urinary polycyclic aromatic hydrocarbons (PAHs) (C) for those reporting not having been told they have bronchitis and those having bronchitis (Selman & Pardo 2012;Olsen et al. 2008;Lantz et al. 2009;Choudhury et al. 2016;Sanchez et al. 2018). This supports our findings of no association between chronic bronchitis and arsenic. ...
Article
Full-text available
Metals, arsenic, and polycyclic aromatic hydrocarbons (PAHs) have all been linked to respiratory diseases. Chronic bronchitis, which is a form of chronic obstructive pulmonary disease (COPD), is a major public health concern and source of morbidity and mortality in the US. The purpose of this study was to analyze the correlation of 14 urinary metals (antimony, barium, cadmium, cesium, cobalt, lead, manganese, mercury, molybdenum, strontium, thallium, tin, tungsten, uranium), seven species of arsenic, and seven forms of polycyclic aromatic hydrocarbon (PAH) concentrations and chronic bronchitis in the US population. A cross-sectional analysis using three datasets from the National Health and Nutrition Examination Survey (NHANES) between 2011 and 2016 in adults, aged 20 years and older. Chronic bronchitis was determined using a self-questionnaire from the NHANES dataset. A specialized weighted complex survey design analysis package was used to analyze NHANES data. Multivariate logistic regression models were used to determine the correlation between urinary metals, arsenic, PAHs, and chronic bronchitis. Models were adjusted for lifestyle and demographic factors. A total of 4186 participants were analyzed; 49.8% were female and 40.5% were non-Hispanic White. All seven types of PAHs showed a positive association with chronic bronchitis (1-hydroxynaphthalene odds ratio (OR): 1.559, 95% confidence interval (CI): 1.271–1.912; 2-hydroxynaphthalene OR: 2.498, 95% CI: 1.524–4.095; 3-hydroxyfluorene OR: 2.752, 95% CI: 2.100–3.608; 2-hydroxyfluorene OR: 3.461, 95% CI: 2.438–4.914; 1-hydroxyphenanthrene OR: 2.442, 95% CI: 1.515–3.937; 1-hydroxypyrene OR: 2.828, 95% CI: 1.728–4.629; 2 & 3-hydroxyphenanthrene OR: 3.690, 95% CI: 2.309–5.896). Of the metals, only urinary cadmium showed a statistically significant positive association (OR: 2.435, 95% CI: 1.401–4.235) with chronic bronchitis. No other metals or arsenic were correlated with chronic bronchitis. Seven forms of urinary PAHs, cadmium, and several demographic factors were associated with chronic bronchitis.
Article
Prenatal arsenic exposure is a major public health concern, associated with altered birth outcomes and increased respiratory disease risk. However, characterization of the long-term effects of mid-pregnancy (second trimester) arsenic exposure on multiple organ systems is scant. This study aimed to characterize the long-term impact of mid-pregnancy inorganic arsenic exposure on the lung, heart, and immune system, including infectious disease response using the C57BL/6 mouse model. Mice were exposed from gestational day 9 till birth to either 0 or 1000µg/L sodium (meta)arsenite in drinking water. Male and female offspring assessed at adulthood (10-12 weeks of age) did not show significant effects on recovery outcomes after ischemia reperfusion injury but did exhibit increased airway hyperresponsiveness compared to controls. Flow cytometric analysis revealed significantly greater total numbers of cells in arsenic-exposed lungs, lower MHCII expression in natural killer cells, and increased percentages of dendritic cell populations. Activated interstitial (IMs) and alveolar macrophages (AMs) isolated from arsenic-exposed male mice produced significantly less IFN-γ than controls. Conversely, activated AMs from arsenic-exposed females produced significantly more IFN-γ than controls. Although systemic cytokine levels were higher upon Mycobacterium tuberculosis (Mtb) infection in prenatally arsenic-exposed offspring there was no difference in lung Mtb burden compared to controls. This study highlights significant long-term impacts of prenatal arsenic exposure on lung and immune cell function. These effects may contribute to the elevated risk of respiratory diseases associated with prenatal arsenic exposure in epidemiology studies and point to the need for more research into mechanisms driving these maintained responses.
Article
Arsenic (As), a toxic metalloid, primarily originates from both natural and anthropogenic activities. Reports suggested that millions of people globally exposed to high levels of naturally occurring As compounds via inhalation and ingestion. There is evidence that As is a well-known lung carcinogen. However, there has been relatively little evidence suggesting its non-malignant lung effects. This review comprehensively summarises current experimental and clinical studies implicating the association of As exposure and the development of several non-malignant lung diseases. Experimental studies provided evidence that As exposure induces redox imbalance, apoptosis, inflammatory response, epithelial-to-mesenchymal transition (EMT), and affected normal lung development through alteration of the components of intracellular signaling cascades. In addition, we also discuss the sources and possible mechanisms of As influx and efflux in the lung. Finally, current experimental studies on treatment strategies using phytochemicals and our perspective on future research with As are also discussed.
Article
Understanding the health effects of exposures when there is a lag between exposure and the onset of disease is an important and challenging topic in environmental health research. The National Institute of Environmental Health Sciences (NIEHS) Superfund Basic Research and Training Program (SRP) is a National Institutes of Health (NIH) grant program that uses a multidisciplinary approach to support biomedical and environmental science and engineering research. Because of the multidisciplinary nature of the program, SRP grantees are well-positioned to study exposure and latent disease risk across humans, animal models, and various life stages. SRP-funded scientists are working to address the challenge of connecting exposures that occur early in life and prior to conception with diseases that manifest much later, including developing new tools and approaches to predict how chemicals may affect long-term health. Here, we highlight research from the SRP focused on understanding the health effects of exposures with a lag between exposure and the onset of the disease as well as provide future directions for addressing knowledge gaps for this highly complex and challenging topic. Advancing the knowledge of latency to disease will require a multidisciplinary approach to research, the need for data sharing and integration, and new tools and computation approaches to make better predications about the timing of disease onset. A better understanding of exposures that may contribute to later-life diseases is essential to supporting the implementation of prevention and intervention strategies to reduce or modulate exposures to reduce disease burden.
Article
Exposure to dust from active and abandoned mining operations may be a very significant health hazard, especially to sensitive populations. We have previously reported that inhalation of real-world mine tailing dusts during lung development can alter lung function and structure in adult male mice. These real-world dusts contain a mixture of metal(oid)s, including arsenic. To determine whether arsenic in inhaled dust plays a role in altering lung development, we exposed C57Bl/6 mice to a background dust (0 arsenic) or to the background dust containing either 3% or 10% by mass, calcium arsenate. Total level of exposure was kept at 100 μg/m3. Calcium arsenate was selected since arsenate is the predominant species found in mine tailings. We found that inhalation exposure during in utero and postnatal lung development led to significant increases in pulmonary baseline resistance, airway hyper-reactivity, and airway collagen and smooth muscle expression in male C57Bl/6 mice. Responses were dependent on the level of calcium arsenate in the simulated dust. These changes were not associated with increased expression of TGF-β1, a marker of epithelial to mesenchymal transition. However, responses were correlated with decreases in the expression of Club cell protein 16 (CC16). Dose dependent decreases in CC16 expression and increases in collagen around airways was seen for animals exposed in utero only (GD), animals exposed postnatally only (PN) and animals continuously exposed throughout development (GDPN). These data suggest that arsenic inhalation during lung development can decrease CC16 expression leading to functional and structural alterations in the adult lung.
Article
We examined the morphological and molecular consequences of acute in utero exposure to teratogenic concentrations of arsenate. The treatment produced a dose-related increase in neural tube defects, along with a significant alteration in the pattern of gene expression for several transcription factors (creb, Hox 3.1, Pax3, and Emx-1) that were examined using in situ transcription and antisense RNA amplification procedures. On gestational day 9:0, there was a significant delay in the embryos progression through neural tube closure, accompanied by a significant downregulation of Hox 3.1 expression and a significant upregulation of Pax3, Emx-1, and creb. As both Hox 3.1 and Pax3 serve to regulate N-CAM expression, it is possible that abnormalities associated with N-CAM may compromise neural crest cell migration and normal neural tube closure.
Article
Allergic asthma is a heterogeneous disease, the pathology of which is a result of improper immune responses to innocuous antigens. We and others have previously shown that one of the Toll-like receptor (TLR)-7/8 ligands, the synthetic compound S28463 (resiquimod, R-848), is able to inhibit acute allergic asthma in mice. Given that the efficiency of this pharmacologic compound against the smooth muscle mass increase and goblet cell hyperplasia that are characteristic of chronic allergic asthma has not been previously assessed, we investigated the ability of this compound to prevent these aspects of chronic airway remodeling. The impact of S28463 treatment was assessed in a Brown Norway rat model of chronic asthma by histologic, morphometric, and molecular techniques. We demonstrate that treatment with S28463 is able to prevent the development of goblet cell hyperplasia and increases in airway smooth muscle mass, and that this effect is at least partially mediated by inhibiting proliferation of goblet and smooth muscle cells, respectively. Furthermore, we show that the abrogation of airway remodeling is preceded by inhibition of the inflammatory reaction normally occurring in response to allergen challenge in sensitized animals. This inhibition was associated with a reduction of both helper T cell type 1 and type 2 cytokine protein expression in the lungs, demonstrating the potent antiinflammatory effect of this pharmaceutical compound in the context of allergic reactions. Taken together, our results indicate great potential for the use of S28463 as an antiinflammatory therapeutic agent for the management of chronic asthma.
Article
In the present study, we characterize the toxic effects of in utero arsenic exposure on the developing lung. We hypothesize that in utero exposure to inorganic arsenic through maternal drinking water causes altered gene and protein expression in the developing lung, indicative of downstream molecular and functional changes. From conception to embryonic day 18, we exposed pregnant Sprague-Dawley rats to 500 ppb arsenic (as arsenite) via the drinking water. Subtracted cDNA libraries comparing control to arsenic exposed embryonic lungs were generated. In addition, a broad Western blot analysis was performed to identify altered protein expression. A total of 59 genes and 34 proteins were identified as being altered. Pathway mapping and analysis showed that cell motility was the process most affected. The most likely affected pathway was alteration in integrin signaling through the beta-catenin pathway, altering c-myc. The present study shows that arsenic induces alterations in the developing lung. These data may be useful in the elucidation of molecular targets and biomarkers of arsenic exposure during lung development and may aid in understanding the etiology of arsenic induced adult respiratory disease and lung cancers.
Article
The lung activity of the antioxidant enzymes (AOEs) copper, zinc superoxide dismutase (Cu,Zn SOD), catalase (CAT), and glutathione peroxidase (GP), but not manganese superoxide dismutase (Mn SOD), increases in rats during late gestation; the concentrations of Cu,Zn SOD mRNA and CAT mRNA also rise. During early postnatal exposure to > 95% O2, the lung activity of Cu,Zn SOD, CAT, and GP increases. We now show 1) the lung concentration of Mn SOD mRNA and GP mRNA does not increase in late gestation; 2) Mn SOD activity and the concentration of its mRNA and of GP mRNA increase during exposure of neonatal rats to > 95% O2; and 3) as previously shown for CAT mRNA, the increase in lung concentration of the mRNAs for Cu,Zn SOD, Mn SOD, and GP during early postnatal hyperoxia occurs with a 70-80% prolongation of the half-life of these mRNAs. We conclude that 1) in late gestation the level at which lung AOE gene expression is regulated differs among the enzymes, 2) the level at which lung AOE gene expression is regulated shortly after birth in response to > 95% O2 is uniform among the enzymes, and 3) the lung's AOE response to neonatal hyperoxia is not merely a step-up of its prenatal regulation but involves different regulatory mechanisms based on increased stability of AOE mRNAs.
Article
Viral bronchiolitis in human infants has been associated with permanent changes in small airways and gas exchange and an increased incidence of hyperresponsive airways later in life. Respiratory infection by Sendai virus in neonatal rats also has been reported to cause permanent changes in lung morphology and increased numbers of bronchiolar mast cells and eosinophils. We evaluated pulmonary mechanics, gas exchange, and airway responsiveness in rats at 7 and 13-16 wk after neonatal Sendai virus infection. Rats from the virus group had lower arterial PO2 and increased total lung resistance compared with controls. There were no significant differences between groups for arterial PCO2, dynamic lung compliance, quasi-static respiratory system compliance, or vital capacity. Rats from the infected group were significantly more sensitive to aerosolized methacholine than were controls, although both virus and control groups became less sensitive with age. We conclude that neonatal Sendai virus infection in rats results in persistent alterations in lung function and airway responsiveness. This phenomenon may be valuable for the study of the relationships among airway inflammation, lung morphology, and airway hyperresponsiveness, and it may be relevant to human airway disease.
Article
Alveolarization of the immature lung is thought to be influenced by the presence of elastic fibers that could provide structural support for developing septa. Although morphometric studies have established that alveolar septal development occurs from days 4 to 13 in the neonatal rat, the precise time period over which elastin synthesis occurs has proved difficult to determine. We have evaluated the usefulness of in situ hybridization techniques to follow tropoelastin message expression in parenchymal tissue, small vessels, and bronchioles in the developing rat lung from days 4 through 18. This method proved to be sufficiently sensitive to detect differences in rates of tropoelastin message expression from days 4 through 18 (P less than 0.0001). Peak tropoelastin message expression was observed in the small vessels on day 4 and in parenchymal tissue on days 9 through 11. Because the time course of tropoelastin message expression in small vessels differs from that in parenchymal tissue, the use of lung extracts to analyze rates of tropoelastin synthesis in the developing lung may be in error.
Article
Age-adjusted mortality rates were analyzed to examine the dose-response relation between ingested arsenic levels and risk of cancers and vascular diseases among residents in the endemic area of blackfoot disease, a unique peripheral vascular disease associated with long-term exposure to high-arsenic artesian well water and confined to the southwestern coast of Taiwan. The arsenic levels in well water determined in 1964-1966 were available in 42 villages of the study area, while mortality and population data during 1973-1986 were obtained from the local household registration offices and Taiwan Provincial Department of Health. Age-adjusted mortality rates from various cancers and vascular diseases by sex were calculated using the 1976 world population as the standard population. A significant dose-response relation was observed between arsenic levels in well water and cancers of the bladder, kidney, skin, and lung in both males and females, and cancers of the prostate and liver in males. However, there was no association for cancers of the nasopharynx, esophagus, stomach, colon, and uterine cervix, and for leukemia. Arsenic levels in well water were also associated with peripheral vascular diseases and cardiovascular diseases in a dose-response pattern, but not with cerebrovascular accidents. The dual effect of arsenic on carcinogenesis and arteriosclerosis and the interrelation between these two pathogenic mechanisms deserve more intensive study.