Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2021 Sep 3;16(9):e0249442. doi: 10.1371/journal.pone.0249442

Prenatal cadmium exposure does not induce greater incidence or earlier onset of autoimmunity in the offspring

Jamie L McCall 1, Harry C Blair 2,3, Kathryn E Blethen 1, Casey Hall 1, Meenal Elliott 1, John B Barnett 1,4,*
Editor: David M Lehmann5
PMCID: PMC8415597  PMID: 34478449

Abstract

We previously demonstrated that exposure of adult mice to environmental levels of cadmium (Cd) alters immune cell development and function with increases in anti-streptococcal antibody levels, as well as decreases in splenic natural regulatory T cells (nTreg) in the adult female offspring. Based on these data, we hypothesized that prenatal Cd exposure could predispose an individual to developing autoimmunity as adults. To test this hypothesis, the effects of prenatal Cd on the development of autoimmune diabetes and arthritis were investigated. Non-obese diabetic (NOD) mice were exposed to Cd in a manner identical to our previous studies, and the onset of diabetes was assessed in the offspring. Our results showed a similar time-to-onset and severity of disease to historical data, and there were no statistical differences between Cd-exposed and control offspring. Numerous other immune parameters were measured and none of these parameters showed biologically-relevant differences between Cd-exposed and control animals. To test whether prenatal Cd-exposure affected development of autoimmune arthritis, we used SKG mice. While the levels of arthritis were similar between Cd-exposed and control offspring of both sexes, the pathology of arthritis determined by micro-computed tomography (μCT) between Cd-exposed and control animals, showed some statistically different values, especially in the female offspring. However, the differences were small and thus, the biological significance of these changes is open to speculation. Overall, based on the results from two autoimmune models, we conclude that prenatal exposure to Cd did not lead to a measurable propensity to develop autoimmune disease later in life.

Introduction

Cadmium (Cd) is a heavy metal and carcinogen present in high levels at battery manufacturing facilities, around zinc smelters, and in cigarette smoke. Cd has been classified as a carcinogen based on epidemiological studies showing a link between Cd exposures with lung and breast cancers [1]. Humans absorb Cd either by ingestion or inhalation [2]. Recent studies on the urinary levels assayed as part of the NHANES reported overall Cd levels of 0.24 μg/g; however, levels of 0.41 μg/g were reported in smokers [3]. Urine Cd levels dropped by 34.3% in the interval between 1988–94 and 2003–2008, which is likely attributable to reduction in smoking [3]. The biological half-life of urinary Cd in the human body using a two-compartment model was 75–128 days for the fast compartment and 7.4–16 years for the slow compartment [4]. Additional studies using a larger cohort and a one-compartment model showed the urinary Cd half-life is around 12 years because of lack of effective Cd elimination pathways [5]. One of the organs with more Cd-induced toxicity is the thymus, the primary site of T-cell production [6]. Pharmacokinetic studies have shown that Cd does not readily reach the fetus in humans, but it accumulates in high concentrations in the placenta [7]. In humans, long-term Cd exposure in utero correlates with hypermethylation of genes in offspring cord-blood when compared to maternal leukocytes [8]. Additionally, it was recently reported that chronic exposure to Cd induces differential methylation in mice spermatozoa [9]. Taken together, these data indicate that there is likely an epigenetic mechanism by which the effects of Cd exposure can be transmitted to the offspring.

Previous studies from our group demonstrate that exposure of adult mice to environmental levels of Cd alters the immune cell development and function of their offspring [1012]. In these studies, prenatal Cd exposure increased the number of CD4+ cells and a subpopulation of double-negative cells (DN; CD4-CD8-), DN4 (CD44+CD25-) in the thymus at birth. These observations correlated with decreases in gene expression of both sonic hedgehog and Wnt/β-catenin [10]. Additionally, we assessed the ability of the 7-week-old offspring of Cd-exposed mice to respond to immunization with a heat-killed Streptococcus pneumoniae (HKSP). HKSP has two immunodominant antigens, pneumococcal surface protein A (PspA), which is a T-dependent antigen, and phosphorylcholine (PC), which is a type 2 T-independent antigen [13,14]. These studies revealed that prenatal Cd-exposure led to a >600-fold average increase in the IgG anti-PspA levels in females and about a 17-fold increase in males [11]. Anti-PC antibody levels also increased in these animals but in a less dramatic manner [11]. In contrast, splenic natural regulatory T cells (nTreg) were decreased in the female offspring [11]. Male offspring did not show a reduction in splenic nTreg cells [11]. At 20 weeks-of-age, offspring of Cd-exposed mice also exhibited persistent changes to thymus and spleen cell phenotypic repertoire, including decreased splenic nTreg cells in both sexes, as well as the adaptive immune response against HKSP [12].

The potential role of Cd in type 1 diabetes has been demonstrated in several experimental and epidemiological studies. Both acute and sub-chronic exposures to Cd have been shown to induce diabetogenic effects in animal models [1517]. Additionally, Cd exposure during pregnancy correlates with an increased risk of gestational diabetes [18]. However, these studies assessed diabetes in humans and animals that were directly exposed to Cd. Given the reported direct effects of Cd on diabetes, increases in IgG antibody to HKSP, and the decreased nTreg cells in prenatally exposed females [11], we formulated the hypothesis that prenatal Cd may provide the initial immune cell changes that would lead to the development of clinical autoimmune disease as adults.

The foundation of our hypothesis was the reported role of nTreg and inducible regulatory T cells (iTreg) in autoimmune disease [1925]. It is also apparent that the role of Treg cells in autoimmune disease is not necessarily correlated with just reduced numbers of Treg cells and that there are likely phenotypic differences, possibly caused by epigenetic changes [2528]. However, to perform mechanistic studies on the effects of prenatal Cd on autoimmunity, whether due to changes in Treg cell numbers or function, requires an animal model in which an outcome can be reproducibly attributed to the treatment. Thus, we sought to develop a model of Cd-induced autoimmunity by investigating the effects of prenatal Cd exposure using two well-established autoimmune animal models: the non-obese diabetic (NOD) mouse and the SKG Balb/C arthritis model, more recently established by Sakaguchi and coworkers [29]. Our models suggest that prenatal Cd exposure does not have a strong in utero effect on the development of autoimmune diseases in the offspring. This report provides guidance for other investigators in the pursuit of the goal of investigating the role of environmental exposures on autoimmune disease.

Materials and methods

Breeding and Cd exposure

All animal procedures were approved by the WVU Institutional Animal Care and Use Committee. Animals were euthanized by cervical dislocation as approved by the committee. Mice are housed in a specific pathogen-free (SPF) barrier facility with a 12 h light/dark cycle. Mice are fed normal chow (Envigo, 2018 Tekland Rodent Diet) and are allowed water ad libitum.

Non-obese diabetic (NOD/ShiLtJ; 001976) mice at 8–10 weeks of age were obtained from Jackson Labs (Bar Harbor, ME). Briefly, after a one-week (minimum) acclimation in our vivarium, 2 females were placed in a cage with one male for 5 days to maximize pregnancy rate. Dams used as controls had free access to normal water and Cd-treated dams had free access to 10 ppm of CdCl2 (Sigma-Aldrich, C3141; ≥ 98% purity) dissolved in water. Briefly, CdCl2 was reconstituted at 101.6 mg/L (5000X) in ddH2O. Stock CdCl2 was diluted to 10 ppm in ddH2O, mixed thoroughly, and transferred to water bottles prior to autoclaving. To reduce the number of animals used for the study, only one dose was used at a level that would reproduce human environmental exposures, as described in the Introduction. The dose of 10 ppm was based on our previous studies which demonstrated immunomodulatory effects in the offspring of mice exposed to Cd [1012]. Previous assessment of water consumption with or without Cd-spike, indicated no difference in water intake between groups [10]. Cd administration was stopped at birth. All offspring were weaned at 3–4 weeks and the dams were euthanized by CO2 asphyxiation. Animals tested for the results reported herein were treated in an identical manner as a cohort tested at birth. In a previous cohort, Cd levels were assessed in the dams and representative offspring at birth using inductively coupled plasma optical emission spectrometry (ICP-OES) as reported in [10]. Cd levels in the kidneys of dams at the time of weaning the litter was 4.37±0.76 μg/g tissue and Cd levels were slightly above the level of detection by the ICP-OES (2.5 ppb) in the liver of offspring when tissues from ≥3 animals were pooled [10]. In this study, offspring at 18–22 weeks of age were euthanized by cervical dislocation and spleens were removed. The 18-week time point would approximate a 25-year-old human with a fully developed immune system. Mice were exposed to Cd in independent cohorts separated by time. Data are presented as combined data.

“SKG mice,” discovered by Sakaguchi et al., are Balb/C mice that have a natural mutation in the Zap70 gene encoding a component of the T cell receptor [30,31]. Raised under SPF conditions, these animals do not spontaneously develop rheumatoid arthritis (RA), but RA can be induced, as described below, and thus, a breeding colony can be maintained. SKG breeding pairs (a generous gift from Dr. Holly Rosenzweig, Oregon Health and Science University) were started with access to water with or without cadmium (10 ppm). Males were removed on day 5 and females were continued on cadmium-spiked or unspiked (control) water until pups were born. Cadmium bottles were exchanged with regular, unspiked water within 12 hours of the birth of pups. Offspring were weaned at day 28 postnatal. Two experimental cohorts were separated by time. Arthritic indices data are presented as summarized data.

Autoimmune assays

Onset of diabetes in the NOD mice was determined by measuring urine glucose levels using the Keto-Diastix (Bayer USA, Whippany, NJ) urine analysis test strips. Weekly measurements were performed starting at 5 weeks and continuing through 18 (females) or 22 (males) weeks and the time-to-disease was noted for each animal.

RA was induced in the SKG SPF mice by intraperitoneal (IP) injection of 2 mg zymosan A (Sigma-Aldrich, St Louis MO; Z4250). These animals develop arthritis within 2–3 weeks of the zymosan A injection. Animals develop symptoms typical of arthritis, which includes swollen joints. An arthritic index (AI) is determined by scoring each paw individually on a scale of 0–4: 0 –normal; 1 –mild, but definite redness and swelling limited to individual digits; 2 –two swollen joints/digits; 3 –three or more swollen joints/digits; 4 –severe swelling of all joints/digits. All paws were evaluated, so that the maximal AI per mouse was 16. To alleviate pain, arthritic mice were given moist chow on a petri dish on the cage floor. Mice with severe arthritis were given soft bedding.

Micro-CT

Following euthanasia, skeletons were fixed overnight in 10% formalin and stored in 70% ethanol at 4°C until needed. Micro-computed tomography (μCT) was performed on a SkyScan 1272 Micro-CT (Bruker, Billerica MA). Images were processed and analyzed in accordance to standard methods [32]. Briefly, the left rear limbs of each animal were scanned in 70% ethanol using the following parameters: isometric voxel size 7 μm, tube voltage (x-ray tube potential) 60 kV, tube current 166 μA, exposure time 650 ms, aluminum filter 0.25mm. The cross-section images were reconstructed using NRecon (v 1.7.1.0; Bruker) with a 20% correction to reduce beam hardening and ring artifacts applied where appropriate. Segmentation was performed using local adaptive thresholding of the trabecular bone to reduce threshold biasing. The region of interest (ROI) was defined in CT Analyzer (v 1.16.9.0+, Bruker). Using the distal epiphysis (growth plate) as a reference point, the ROI was offset 245 μm (35 slices) proximal to ensure that no growth plate was included in the analysis of the metaphyseal trabecular bone. A total of 1.05 mm (150 slices) was used for quantitative analyses. Trabeculae were delineated from cortical bone using a manual assessment every 15–20 slices. Bone mineral density (BMD) was measured at the distal femur using the same Volume of Interest (1.05 mm) following calibration with calcium hydroxyapatite phantoms of known density: 0.25 and 0.75 g/cm3. Phantoms were prepared, scanned, and reconstructed with the same parameters as above. Three dimensional images were prepared using DataViewer (v 1.5.6.2, Bruker) and CTVOX (3.3.0r1403, Bruker). Images representing the median mouse of each group are presented.

Tissue isolation and cell preparation

Spleens were harvested from euthanized mice and single cell suspensions were prepared: spleens were dissected immediately after euthanasia and submerged into 5 ml of ice-cold phosphate-buffered saline without calcium or magnesium (PBS; Corning, 21-031-CV). Spleens were smashed between frosted sections of sterile microscope slides and passed through a 20G needle 3–4 times to obtain single-cell suspensions. Cells were washed once with PBS and red blood cells were lysed using RBC lysis buffer (Sigma-Aldrich, R7757). After neutralization with complete medium [RPMI-1640 medium (Corning, 15-040-CV) supplemented with 10% heat-inactivated fetal bovine serum (FBS, Sigma-Aldrich, F0926), L-glutamine (2 mM; Gibco, 25030–081), HEPES (5mM; Gibco, 15630–080), penicillin/streptomycin (100 units/L and 100 μg/ml; Cellgro, 30-002-C1), and 2-mercaptoethanol (0.05 mM, Sigma-Aldrich, M3148)], cells were washed once in staining buffer consisting of PBS pH 7.5, 0.5% bovine serum albumin (BSA; Sigma-Aldrich, A7030), and 2 mM ethylenediaminetetraacetic acid (EDTA; Sigma-Aldrich, E5134) for flow cytometry staining or complete medium for cell culture stimulation. Viable cells were enumerated using trypan blue and a hemocytometer. Cell preparations from each mouse were analyzed individually for cytokines and splenocyte populations (see below).

Primary cell culture and anti-CD3/28 stimulation

Splenocytes were stimulated with anti-CD3 and anti-CD28 in vitro. Briefly, 96 well tissue culture plates were coated with 50 μL PBS (with calcium and magnesium, Corning, 21-030-CV) or 5 μg/mL anti-CD3 (clone 17A2, eBioscience, 14-0032-86) diluted in PBS. Plates were incubated ~3 hours at 37°C. Wells were washed twice with 200 μL PBS, cells were diluted to 1x106/mL, and 200 μL of cells were placed in 5 wells per mouse per treatment (5 PBS, 5 anti-CD3). Cells were stimulated by the addition of anti-CD28 (clone 37.51, eBioscience, 14-0281-86) at 2 μg/mL final concentration and incubated for 72 hours at 37°C with 5% CO2. Supernatants were collected and stored at -80°C until assay. Cell supernatants from each mouse were analyzed individually.

Cell staining and flow cytometry

Single cell suspensions of splenocytes were prepared as described above. Surface and intracellular staining was performed as follows. Splenocytes were stained with either a cocktail of antibodies against CD3, CD19, CD4, CD8 (staining set 1) or CD4, CD25, and FOXP3 (staining set 2). All antibodies were used at a 1:200 dilution. A detailed list of staining panels, including antibody clones, fluorochrome tags, and the source of each antibody is provided in Table 1. Briefly, cells (1 x 106) were washed with staining buffer and incubated with whole rat and mouse IgG (Jackson ImmunoResearch, West Grove, PA) for 30 min on ice to block Fc receptors. Without washing, cells were stained with LIVE/DEAD Fixable Yellow Dead Stain kit (Invitrogen, L34959) at a dilution of 1:2000 for 30 minutes on ice in the dark. Cells in both staining sets were washed once in staining buffer, incubated for 30 min on ice with fluorochrome-labeled antibodies to detect surface markers followed by several washes with staining buffer. For staining set 1, stained cells were fixed overnight at 4°C with 0.4% paraformaldehyde. Paraformaldehyde was removed by washing with PBS and final resuspension in PBS. Fixed cells were stored at 4°C until analyzed. For intracellular staining (staining set 2), cells were fixed and permeabilized using the eBioscience™ FOXP3/Transcription Factor Staining Buffer Set (Invitrogen, 00-5523-00) and stained with anti-FOXP3 antibody according to the manufacturer’s protocol.

Table 1. Antibodies used for spleen phenotyping.

Staining Panel 1
Target Clone Fluorochrome Manufacturer Catalog #
CD3 145-2C11 FITC BD Pharmingen 553062
CD4 RM4-5 APC-eFluor 780 Invitrogen 47-0042-82
CD8 53–6.7 V450 BD Horizon 560489
CD19 1D3 PE BD Pharmingen 557399
Staining Panel 2
Target Clone Fluorochrome Manufacturer Catalog #
CD4 (surface) RM4-5 APC-eFluor 780 Invitrogen 47-0042-82
CD25 (surface) 7D4 PE BD Pharmingen 558642
FOXP3 (intracellular) FJK-16S Efluor450 eBioscience 48-5773-82

Data were acquired using a BD LSRFortessa and Diva 8.0 software (BD Biosciences) and analyzed using FCS Express software (De Novo Software, San Jose, CA). A minimum of 10,000 events were collected for each sample. Gating strategies are shown in S1 Fig.

Cytokine analysis

Cell culture supernatants from the anti-CD3/CD28 stimulated splenocytes were analyzed for cytokine concentrations using MSD kit U-Plex Biomarker Group 1 (ms) Assays (Meso Scale Diagnostic, K15069L-1) with 9 analytes (GM-CSF, IL-17A, IL-17F, IL-1β, IL-21, IL-23, IL-6, KC/GRO, and TNF-α) according to the manufacturer’s protocol. Samples were diluted 1:2 prior to addition to the plate. Plates were read on the MSD HTS24 plate reader.

Anti-nuclear antibody screening

Sera from SKG mice were assessed for the presence of anti-nuclear antibodies (ANA) using the ANA Hep Screen ELISA Kit (Abnova, KA1080, lot 2106180). This assay is a screening system to qualitatively measure IgG class autoantibodies against SS-A-52 (Ro-52), SS-A-60 (Ro-60), SS-B (La), RNP/Sm, RNP-70, RNP-A, RNP-C, Sm-bb, Sm-D, Sm-E, Sm-F, Sm-G, Scl-70, Jo-1, dsDNA, ssDNA, polynucleosomes, mononucleosomes, histone complex, histone H1, histone H2A, histone 3, histone H4, Pm-Scl-100, and centromere B. Methodology was adapted to rodent studies by replacing the secondary antibody with HRP-conjugated goat anti-mouse IgG (BD Phamingen, Cat 554002, lot 5247553) in which is consistent with previous publications using similar technologies [33].

Statistical analyses

All statistics were performed using GraphPad Prism version 8. Time to disease curves were compared within each sex using a Log-rank (Mantel-Cox) test and diabetes incidence was analyzed using Fisher’s Exact test. When comparing two groups (e.g. Cd versus Control offspring), data were analyzed using two-tailed student’s t-tests. When analyzing four groups (e.g. stratified by disease incidence), data were analyzed using an ANOVA with Tukey’s post-hoc test for multiple comparisons. * p<0.05, ** p<0.01, *** p<0.001. Raw data used to generate graphs presented as means are included in S1 Table.

Results

Incidence of diabetes in NOD offspring

To determine the effect of prenatal Cd exposure on autoimmune disease, the incidence and time-to-disease of diabetes was measured in NOD offspring. NOD parents were exposed to Cd as described under Methods and the offspring monitored for elevated glucose as an indicator of diabetes. Female Cd offspring showed a slight delay in the development of diabetes as compared to female control offspring (Fig 1A). This difference was not statistically significant. The percentage of diabetes-positive female offspring and the number of litters composing each group is shown in Fig 1B. Male offspring developed diabetes (both treatment groups) later than the female offspring (Fig 1A). There was essentially no difference in the incidence of diabetes or time of onset between the male Cd offspring and control offspring (Fig 1A and 1B). Thus, by these measures prenatal exposure to Cd did not cause a significant change in autoimmune disease development.

Fig 1. Time to disease and incidence of diabetes of Cd-exposed NOD offspring.

Fig 1

Diabetes incidence was based on elevated glucose in the urine of the offspring at the indicated times. (A) Time (weeks) until a positive urine test. Number of mice per group are indicated in the legend. (B) Percent of offspring in the litters that were diabetic at 18 weeks (females) and 22 weeks (males). Number of dams associated with these litters shown above the bar.

Splenic phenotypes in NOD offspring

Splenic phenotypes were assayed in the offspring at 18 (females) and 22 (males) weeks of age. S2 Fig (S2 Fig) shows the number of CD4+, CD8+ and CD19+ splenocytes in each group. No differences in the numbers of any of these cell phenotypes were seen.

We previously reported decreased numbers of nTreg cells in the Cd-exposed offspring in a C57BL/6 mouse model [11]. Thus, we sought to determine if nTreg cell numbers in the NOD offspring was also altered by prenatal Cd exposure. We first assessed whether prenatal Cd exposure altered Treg percentages, regardless of disease state. As shown in Fig 2, prenatal Cd exposure does not alter nTreg percentages. We hypothesized that offspring with measurable disease would have reduced Tregs in the spleen. However, no differences in nTreg cell number were observed when stratified by disease state in any treatment group of either sex (Fig 3). No changes in total splenocyte numbers were observed between any of the treatment groups for either sex. Therefore, Cd exposure did not cause changes in the numbers of the total cell number, major splenocyte phenotypes, or specifically of the Treg cells.

Fig 2. Analysis for Treg spleen cell percentages of Cd-exposed NOD offspring.

Fig 2

At 18 weeks (females) and 22 weeks (males), the spleens of the offspring were ablated and processed for flow cytometric analysis. CD4+CD25+FOXP3+ were quantified. Data are shown as mean ± SD. Males N = 15–21; Females N = 10–13.

Fig 3. Analysis for Treg spleen cells of Cd-exposed NOD offspring.

Fig 3

At 18 weeks (females) and 22 weeks (males), the spleens of the offspring were ablated and processed for flow cytometric analysis. CD4+CD25+FOXP3+ were quantified. Data are shown as mean ± SD. Males N = 5–13; Females N = 4–9.

Splenic cytokine production in NOD offspring

Several cytokines are associated with the development of autoimmune disease [34,35]. These are predominantly the cytokines that contribute to inflammation, and include IL-17A and IL-17F as well as TNF-α. In male offspring, at 22 weeks there was a statistically significant decrease in IL-17F between the control diabetic and the Cd-exposed diabetic and non-diabetic animals (Fig 4); however, whether these differences were biologically significant is not obvious. No differences were seen in either IL-17A or TNF-α levels in these male offspring.

Fig 4. Spleen cell proinflammatory cytokine production by Cd-exposed NOD offspring.

Fig 4

At 18 weeks (females) and 22 weeks (males), the spleens of the offspring were ablated and stimulated with anti-CD3+anti-CD28 for 72h. Cytokine levels were measured using MSD multiplex plates. Symbols represent the data for individual mice and the black bar denotes the median. Males N = 5–17; Females N = 4–14.

Similar assays were performed on the female offspring at 18 weeks. IL-17A was reduced in non-diabetic female Cd animals as compared to non-diabetic controls (<2-fold, p<0.05), indicating a possible inhibiton of inflammation with prenatal Cd exposure in the female offspring (Figs 4 and S3). Again, the biological significance of this difference is not obvious (Fig 4). No significant differences were noted in the cytokine production levels of any of the other cytokines shown in Fig 4.

Additional cytokines were also measured in the spleen cells from these offspring. These included IL-1β, IL-21, IL-23, IL-6 and KC/GRO (S3 Fig). No significant differences were noted in the ability of splenocytes to produce these cytokines were seen in either sex regardless of treatment group.

Arthritis incidence in the SKG mouse strain

The specific-pathogen-free (SPF) reared SKG mice develop arthritis after zymosan A injection. At 8 weeks, arthritis was induced in male and female SKG offspring to determine if differences in the arthritis incidence or severity is due to prenatal Cd exposure. Arthritic index (AI), which is a measure of arthritic-inflammation in the paws was assessed weekly starting one week post zymosan A-induction. A positive AI score (as further described in the Methods) is indicated by visible swelling in one or more joint within the paw. Each paw is measured on a scale of 1–4 with a total possible score of 16 per animal (4 paws). No differences in AI between the treatment groups in either sex was apparent (Fig 5). However, females, regardless of treatment, showed a positive AI 3 to 4 weeks earlier than males. By this measure, prenatal Cd exposure did not cause an increase in arthritic inflammation.

Fig 5. Incidence of arthritis in Cd-exposed SKG offspring.

Fig 5

At 8 weeks, the arthritis was induced in the offspring of control and Cd-exposed parents by the injection of zymosan A. The arthritic index was determined in the over a 12-week period of time. Data from two independent cohorts were combined are presented as mean ± SEM. Number of mice per exposure group are indicated in the legend.

Direct Cd exposure via drinking water has been shown to induce the presence of antinuclear antibodies (ANA) in the blood of mice [33]. To assess the presence of ANA in the offspring of exposed mice we screened for autoantibodies in the sera using an ELISA method adapted for rodents. A single male mouse exhibited autoantibodies above background (S4 Fig); however this did not correlate with increased disease, i.e. arthritic index.

Microcomputed tomographic analysis of the femurs of arthritic SKG mice

In our experience, the AI does not accurately reflect possible bone erosion that accompanies arthritis. Micro-CT (μCT) has been considered the gold standard for assessing trabecular bone microarchitecture [36,37]. For this reason, μCT visualization and analyses were also conducted on the paws of these SKG mice. Fig 6 shows a typical sample from representative animals of all experimental groups. As noted in Fig 6B and 6D, two representatives from the male and female cohorts displayed essentially identical AI scores. Visible in the x-ray scan of the Cd female (Fig 6A) is an obvious distortion (indicated by an arrow) in the phalanges of the paw. This distortion is not present in the control female; however, it is not clear whether this was due to increased brittleness and distortion caused by in vitro experimental manipulation or represents a true bone defect. This distortion was not seen consistently in all Cd female μCT scans. No other differences in the μCT x-ray scans were noted in male offspring, of either treatment group.

Fig 6. MicroCT analysis of the paws of arthritic Control and Cd-exposed SKG offspring.

Fig 6

Dam and sire SKG mice were exposed to cadmium as described. At 8 weeks, the arthritis was induced in the offspring by the injection of zymosan A. Twelve weeks after arthritis induction, the animals were euthanized, and the limbs processed for μCT analysis. (A & C) Representative μCT images from rear left paws. Distorted phalanges are indicated by the arrow. (B & D) Arthritic indices for individual paws shown in A & C. No arthritis control values are inidicated by the dotted line.

Quantitative morphometry was performed on representative μCT images of the femurs from each experimental group and the results are shown in Fig 6. The volume of interest (VOI) was defined as described in Micro-CT methods. Representative images from each sex and treatment group are illustrated in Fig 7A, with the VOI highlighted in red. Several parameters were assessed: 1) percent bone volume (BV/TV) which quantifies the percent of space occupied by trabecular bone within the VOI, 2) trabecular number is also known as the spatial density and determined by the number of times per mm that random line in the VOI crosses a trabecula, 3) trabecular thickness is the average thickness as measured by virtual sphere-fitting, 4) trabecular separation is the diameter of spaces between the trabeculae as assessed using 3D sphere fitting, 5) bone surface to bone volume ratio (BS/BV) which is a general indicator of structural complexity and inversely related to trabecular thickness, and 6) bone surface to total VOI ratio (BS/TV) which quantifies the bone surface density. The Cd females showed a statistical difference in the bone volume fraction (BV/TV) and trabecular thickness (Fig 7B)–values that have been deemed crucial for determining the three-dimensional (3D) outcomes for trabecular bone microarchitecture [36]. The ratio of the segmented bone surface to the segmented bone volume (BS/BV; Fig 7) was also statistically different in the female Cd-exposed offspring. Male offspring showed no statistical differences in any assessment of the trabecular bone architecture (Fig 7).

Fig 7. MicroCT analysis of the femurs of arthritic Control and Cd-exposed SKG offspring.

Fig 7

Femurs of control and Cd-exposed offspring were analyzed by μCT. (A) Cross sections of μCT images of mice representing median mice presented in B. The VOI is highlighted red and shown enlarged below. (B) Quantification of trabecular bone architecture. BV/TV–bone volume/total volume of interest, BS/TV–bone surface/total volume of interest, BS/BS–bone surface/bone volume. Data are presented as mean ± SD. N = 4 per group.

BMD was assessed in the femurs of male and female offspring following prenatal Cd exposure. Values for the no arthritis control mice are indicated by the dotted line. BMD was significantly increased in the femurs of Cd-exposed female offspring as compared to the controls (S6 Fig). No differences were observed in the male offspring.

Discussion

Autoimmune disease is a topic of intense interest because of its life-changing consequences. Treg cells are widely regarded as the key to preventing autoimmune disease because of their well-established role of controlling immune responses and a plethora of studies that have established a potential role of Treg cells in autoimmunity development. In humans, one possible scenario is that some defining initiating event alters Treg cells such that subsequent event(s), presumably separated by time, precipitate alteration in numbers and/or functions of Tregs in a manner that results in autoimmunity. Often neither the initial defining event(s), nor the triggering event(s) are known. It is also too simplistic to attribute autoimmunity solely to the aberrant actions of the Treg cell populations. In addition, heterogeneity of responses by humans to the same stimuli makes these key events increasingly difficult to define.

Exposure to environmental xenobiotics has been postulated as a possible initiating event but may also later trigger frank autoimmune disease [33]. There are a wide range of environmental agents that have been suggested as possible triggers [38]. There are several indicators that Cd may be involved in developing susceptibility to autoimmune disease. For example, direct Cd exposure of adults has been implicated in diabetes development (reviewed in [39]). A relatively unexplored exposure paradigm is the possible epigenetic changes induced in the developing fetus when the parents are exposed to an environmental agent. In this scenario, the environmental agent would be considered the initiating event with the subsequent triggering event undefined. Our previous reports show that prenatal Cd exposure altered several key immune parameters in the offspring suggesting that this treatment could be the initiating event possibly leading to development of autoimmune disease. Based on these data, we embarked on studies to develop a reliable animal model that would allow us to study the role of Cd in prenatally-induced autoimmune susceptibility.

The first autoimmunity model that we tested was the NOD mouse model. NOD mice have been extensively used to investigate the mechanism of autoimmune-induced diabetes and given the previous reports of Cd inducing diabetes with direct exposure, it seemed like a reasonable model to explore. There are numerous reports linking the onset of diabetes in the NOD mouse to functional changes in Treg cells, which prompted us to hypothesize that prenatal exposure of NOD mice to Cd might lead to acceleration of diabetes as in other model systems [40]. Our findings on time-to-onset in NOD mice agree with published studies [40]. Based on the incidence of diabetes, females showed diabetic symptoms earlier and in a higher percentage than male counterparts. Within the female cohort, Cd-exposed offspring generally did not show signs of diabetes until approximate 2 weeks later than the control group; however, once they began to show diabetes, the Cd-exposed group quickly caught up with the controls (Fig 1A). Whether this delay in onset and more rapid progression of the Cd-exposed group has biological significance is unknown. However, we found no statistically significant differences in our studies. Males, in contrast, showed similar onset timing and incidence regardless of treatment group.

In this study we found that prenatal Cd exposure did not alter splenocyte cell populations; this is in contrast to our previous publication where prenatal Cd exposure was associated with reduced splenic Tregs in female offspring and increased splenic Tregs in male offspring [11]. Of note, the previous study examined the effects in offspring using a C57BL/6 background. Prenatal Cd exposure may not induce the same changes in nTreg percentages and overall numbers in the NOD mouse model which has a NOD/ShiLtJ background. Although there is a large body of literature designating the defect in NOD mice as Treg mediated, there is also a report that the defect in NOD is in the T-effector (Teff) cell population [41]. Therefore, a lack of a change in diabetes in this mouse model, may not preclude a possible defect in the Treg population. The effect of prenatal Cd exposure on Treg suppressive function should be assessed in future studies.

To assess T cell function, we stimulated splenocytes in vitro with anti-CD3/anti-CD28 and measured proinflammatory cytokines secreted in the media (Figs 4 and S3). We predicted that prenatal Cd exposure would increase IL-17 levels in the sera. However, we observed that IL-17A was reduced in non-diabetic female Cd animals as compared to non-diabetic controls and IL-17F levels were lower in male Cd-exposed (diabetic and non-diabetic) offspring than in control diabetic animals. However, the data are very noisy and do not engender confidence that these differences have any biological significance, especially when IL-17 levels were decreased in the Cd-exposed offspring. Previous studies in the NOD model show that IL-17A and IL-17F expression correlates with increased autoimmune-mediated type I diabetes [42] and pharmacological inhibition of Th17 cells prevented the development of diabetes in these mice [43]. Conversely, IL-17 deficient NOD mice only showed delayed onset of diabetes and did not result in decreased cumulative incidence in this model [44]. Taken together, we would predict that diabetic mice in our study would have increased IL-17 levels as compared to the controls. However, we did not observe differences in IL-17 production between non-diabetic and diabetic mice regardless of prenatal exposure. Of note, we measured ex vivo cytokine production of splenocytes. These measurements may not be indicative of IL-17 production in vivo in organs important to the development and progression of type 1 diabetes, such as the pancreas. Based on these observations, we conclude that prenatal Cd exposure did not alter the development of diabetes in NOD mice.

In addition to IL-17, we assessed the levels of TNF-α, IL-1β, IL-21, IL-23, IL-6, and KC/GRO. No differences were observed in the ability of T cells to produce these cytokines upon stimulation. In our previous C57BL/6 studies [11,12], we reported significant reduction in IFNγ production in prenatal Cd offspring; however, we did not measure IFNγ levels in the present study. While IFNγ is generally thought to be important in the development of type 1 diabetes, in the NOD model, the lack of IFNγ or IFNγ receptor does not prevent the development of disease [45,46]. Furthermore, the role of IFNγ in animal models of RA remains controversial as it is shown to be protective in the collagen-induced model but required for arthritis development when induced by glucose-6-phosphate isomerase [47]. However, we acknowledge that a limitation of our study is that we did not assess IFNγ or anti-inflammatory cytokines in these models.

Given the failure to demonstrate that the NOD mouse was a suitable model to study possible prenatal Cd autoimmune induction, we tested a second autoimmune model which presents with arthritis. The SKG mouse arthritis model, identified by Sakaguchi and associates [29], has a mutation in the ZAP70 signaling molecule that was caused by a spontaneous event. The mutation alters developing T cells. Treg cell production and function are affected allowing the development of polyclonal self-reactive arthritogenic T cells [48]. These mice, when raised in SPF conditions typical in modern vivaria, remain disease-free until injected with zymosan A (this report) or mannan [49]. Once induced, animals develop arthritis within 2–3 weeks. This model was chosen over collagen-induced arthritis because the arthritis occurs in SKG females earlier and with greater severity than males, which recapitulates the demography of human arthritis [49]. The Cd and control female offspring showed earlier and slightly more severe arthritis than their male counterparts; however, within a given sex, no statistic or apparent differences were observed (Fig 5).

Previous studies have demonstrated presence of ANA in 10% of female FZBW F1 mice exposed to 10 ppm Cd for 4 weeks [33]. To test if ANA were present in the offspring of mice exposed to Cd water for 21 days, we screened the plasma using an ELISA-based method for IgG class autoantibodies against SS-A-52 (Ro-52), SS-A-60 (Ro-60), SS-B (La), RNP/Sm, RNP-70, RNP-A, RNP-C, Sm-bb, Sm-D, Sm-E, Sm-F, Sm-G, Scl-70, Jo-1, dsDNA, ssDNA, polynucleosomes, mononucleosomes, histone complex, histone H1, histone H2A, histone 3, histone H4, Pm-Scl-100, and centromere B. In contrast to direct Cd exposure, only a single male offspring demonstrated levels above background (S4 Fig) in our study. Prenatal Cd exposure did not cause the reduction in spleen weight observed with the Leffel et al. study [33]. Taken together, these data suggest that prenatal Cd exposure does not elicit the same effects in the offspring as direct Cd exposure does in adult animals. Cd does not cross the placental barrier [7].; therefore, it is not suprising that direct Cd exposure may have a different mechanism of action than indirect exposure in the womb. Further experiments are needed to elucidate the mechanisms by which prenatal Cd exposure drives immunological changes in the offspring.

In other experimental systems used in our laboratory, we have noted that the arthritic index (Fig 5), does not necessarily reflect changes in bone architecture. We therefore employed μCT to investigate bone architecture in control animals and offspring of Cd-exposed animals (Figs 6 and 7). X-ray micrographs from representative animals (Fig 6) do not show any notable differences in the bone architecture of the paw beyond what was previously noted as deformation of the phalanges. Quantitative analysis of femurs (Fig 7) revealed small but statistically significant differences in several parameters within the female offspring cohort, including BV/TV, trabecular thickness, and BS/BV. While statically significant, the numerical difference in the mean trabecular bone thickness in the female offspring is only 5.2 μm which is smaller than one pixel (7 μm). Therefore, this difference is less than the resolution of our instrumentation and whether these differences are real or represent a true biological defect is doubtful. As the BS/BV measurement is inversely proportional to the trabecular thickness, the data presented are deceptive and may not accurately reflect the biology.

Unexpectedly, the percentage of bone volume as compared to the total volume (BV/TV) in our VOI was significantly higher in the prenatal Cd female offspring as compared to the control female offspring. The detrimental effects of Cd on bone development have been previously reported with both direct exposure to cells [5052] as well as prenatal exposure in rats [53]. Low levels of Cd (7.5–60 nM) stimulate osteoclastic differentiation in the presence of osteoblasts [51]. Additionally, Cd induces apoptosis in primary rat [52] and human [50] osteoblasts at concentrations of 1–5 μM and 10–50 μM, respectively. Taken together, these studies demonstrate that direct Cd exposure both promotes bone destruction through osteoclast differentiation and inhibits bone repair through the destruction of osteoblasts.

Prenatal Cd exposure (50 ppm via drinking water) in rats results in decreased femur weight, but not body weight, and length [53]. The authors demonstrate that this was due to the ability of Cd to prevent transfer of essential elements, including zinc, to the fetus. The phenotype was rescued with supplemental zinc in diet of the pregnant dam [53]. In contrast, our data indicate that the female Cd offspring had increased BV/TV as compared to the female controls (albeit similar to male Cd and control offspring); therefore, we cannot reasonably conclude that prenatal Cd exposure is causing the differences observed in our study. In attempt to identify additional explanations for our data, we assessed the weight and limb-specific arthritis in the female offspring. The zymosan A injection which induces RA in this SKG model is a single dose of 2 mg, regardless of animal weight. We determined that the weights of the female offspring at the time of the arthritis induction were not different (S5A Fig) and therefore did not result in the observed differences in bone structure. Further, we know that arthritis development in this mouse model, as well as the collagen-induced arthritis (CIA) model, is asymmetric in the limbs. We hypothesized that the differences observed in our trabecular microarchitecture measurements are purely due to varying degrees of arthritis in the scanned limb. However, we found no differences in AI score among the groups (S5B Fig). Finally, while statistically significant, the BV/TV percentage in female control and Cd offspring are highly variable and the difference in the means is small (5.8%). The true biological significance of this parameter is open to speculation. No differences in trabecular bone architecture were observed in the control and Cd male SKG offspring.

Patients with rheumatoid arthritis have a higher risk of developing low BMD and osteoporosis. BMD was assessed in the femurs of male and female offspring following prenatal Cd exposure. BMD was significantly increased in the femurs of Cd-exposed female offspring as compared to the controls (S6 Fig). Consistent with the microarchitecture measurements above, Cd-exposed female offspring had higher BMD than the control offspring. Of note, the control offspring appear to have more bone erosion and less BMD than any other group. If anything, taken together, these data suggest that prenatal Cd exposure appears to be protective in the development of arthritis in the female offspring using the SKG model. Therefore, we conclude that prenatal exposure to Cd does not predispose or exacerbate autoimmune disease in NOD and SKG mice.

Conclusions

In summary, this report details our attempts to develop a reliable model to test the hypothesis that prenatal Cd exposure contributes to the development of autoimmunity. While there were tantalizing hints of an effect, neither of the two models investigated provided data that would unequivocally denote that prenatal Cd induced a higher propensity for Cd-exposed offspring to develop autoimmune disease. Without a consistent model, mechanistic experiments were not possible. However, the data reported herein highlight the importance of model validation and, we hope, will assist those using mouse models of autoimmunity to investigate the long-term outcomes of prenatal exposures.

Supporting information

S1 Fig. Gating strategies for flow cytometry panels.

(TIF)

S2 Fig. Spleen cell phenotypes of cadmium exposed NOD mice.

Dam and sire NOD mice were exposed to cadmium as described. At 18 weeks (females) and 22 weeks (males), the spleens were ablated and processed for flow cytometric analysis. CD4+, CD8+ and CD19+ were quantified. Symbols represent the data for individual mice and the black bar denotes the mean. Males N = 6–16; Females N = 13–19.

(TIF)

S3 Fig. Spleen cell cytokine production by cadmium exposed NOD mice.

Dam and sire NOD mice were exposed to cadmium as described. At 18 weeks (females) and 22 weeks (males), the spleens of the offspring were ablated and stimulated with anti-CD3+anti-CD28 for 72h. Cytokine levels were measured using MSD multiplex plates. No differences were noted in production of these cytokines between any group. Symbols represent the data for individual mice and the black bar denotes the median. Males N = 5–17; Females N = 4–14.

(TIF)

S4 Fig. Prenatal Cd exposure does not induce ANA in SKG model.

Serum autoantibodies were measured by ANA Hep Screen ELISA.

(TIF)

S5 Fig. SKG mice body weight at time of induction and AI of left rear paws.

A) Body weights of male and female mice at the time of arthritis induction with zymosan A. Symbols represent the data for individual mice and the black bar denotes the median. B) Left rear paw arthritic indices. No differences were noted between control and cadmium exposure within each sex. Data are shown as mean ± SEM.

(TIF)

S6 Fig. BMD analysis of the femurs of arthritic Control and Cd-exposed SKG offspring.

Bone mineral density (BMD) was measured at the distal femur using the same Volume of Interest (1.05 mm) following calibration with calcium hydroxyapatite phantoms of known density: 0.25 and 0.75 g/cm3. Phantoms were prepared, scanned, and reconstructed with the same parameters as Fig 7. Data are shown as mean ± SD.

(TIF)

S1 Table. Raw data used for graphs presented as means.

(XLSX)

Acknowledgments

We thank Michelle Witt for her training in microCT image acquisition and analyses.

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

JBB ES023845. ES023845 National Institute for Environmental Health Sciences www.niehs.nih.gov The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. West Virginia University Flow Cytometry & Single Cell Core Facility OD016165 National Institute of General Medical Sciences https://www.nigms.nih.gov/ The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. Institutional Development Awards (IDeA) GM103488 (CoBRE) and GM103434 (INBRE) https://www.nigms.nih.gov/ The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. West Virginia University Animal Models & Imaging Facility RR016440 and P30 RR032138/GM103488 National Institute of General Medical Sciences https://www.nigms.nih.gov/ The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. West Virginia University Cancer Institute https://wvucancer.org/ The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Nawrot T, Plusquin M, Hogervorst J, Roels HA, Celis H, Thijs L, et al. Environmental exposure to cadmium and risk of cancer: a prospective population-based study. Lancet Oncol. 2006;7(2):119–26. doi: 10.1016/S1470-2045(06)70545-9 [DOI] [PubMed] [Google Scholar]
  • 2.Waalkes MP. Cadmium carcinogenesis. Mutat Res. 2003;533(1–2):107–20. Epub 2003/12/04. doi: 10.1016/j.mrfmmm.2003.07.011 . [DOI] [PubMed] [Google Scholar]
  • 3.Tellez-Plaza M, Navas-Acien A, Caldwell KL, Menke A, Muntner P, Guallar E. Reduction in cadmium exposure in the United States population, 1988–2008: the contribution of declining smoking rates. Environmental health perspectives. 2012;120(2):204–9. Epub 2011/11/09. doi: 10.1289/ehp.1104020 ; PubMed Central PMCID: PMC3279452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Jarup L, Rogenfelt A, Elinder CG, Nogawa K, Kjellstrom T. Biological half-time of cadmium in the blood of workers after cessation of exposure. Scand J Work Environ Health. 1983;9(4):327–31. Epub 1983/08/01. doi: 10.5271/sjweh.2404 . [DOI] [PubMed] [Google Scholar]
  • 5.Ishizaki M, Suwazono Y, Kido T, Nishijo M, Honda R, Kobayashi E, et al. Estimation of biological half-life of urinary cadmium in inhabitants after cessation of environmental cadmium pollution using a mixed linear model. Food Additives & Contaminants: Part A. 2015;32(8):1273–6. doi: 10.1080/19440049.2015.1052573 [DOI] [PubMed] [Google Scholar]
  • 6.Viau M, Collin-Faure V, Richaud P, Ravanat JL, Candeias SM. Cadmium and T cell differentiation: limited impact in vivo but significant toxicity in fetal thymus organ culture. ToxicolApplPharmacol. 2007;223(3):257–66. doi: 10.1016/j.taap.2007.05.017 [DOI] [PubMed] [Google Scholar]
  • 7.Levin AA, Kilpper RW, Miller RK. Fetal toxicity of cadmium chloride: the pharmacokinetics in the pregnant Wistar rat. Teratology. 1987;36(2):163–70. Epub 1987/10/01. doi: 10.1002/tera.1420360203 . [DOI] [PubMed] [Google Scholar]
  • 8.Sanders AP, Smeester L, Rojas D, DeBussycher T, Wu MC, Wright FA, et al. Cadmium exposure and the epigenome: Exposure-associated patterns of DNA methylation in leukocytes from mother-baby pairs. Epigenetics. 2014;9(2):212–21. Epub 2013/10/28. doi: 10.4161/epi.26798 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Saintilnord WN, Tenlep SYN, Preston JD, Duregon E, DeRouchey JE, Unrine JM, et al. Chronic Exposure to Cadmium Induces Differential Methylation in Mice Spermatozoa. Toxicological Sciences. 2021. doi: 10.1093/toxsci/kfab002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hanson ML, Brundage KM, Schafer R, Tou JC, Barnett JB. Prenatal cadmium exposure dysregulates sonic hedgehog and Wnt/[beta]-catenin signaling in the thymus resulting in altered thymocyte development. Toxicology and Applied Pharmacology. 2010;242(2):136–45. doi: 10.1016/j.taap.2009.09.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hanson ML, Holaskova I, Elliott M, Brundage KM, Schafer R, Barnett JB. Prenatal cadmium exposure alters postnatal immune cell development and function. Toxicol Appl Pharmacol. 2012;261(2):196–203. doi: 10.1016/j.taap.2012.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Holaskova I, Elliott M, Hanson ML, Schafer R, Barnett JB. Prenatal cadmium exposure produces persistent changes to thymus and spleen cell phenotypic repertoire as well as the acquired immune response. Toxicol Appl Pharmacol. 2012;265(2):181–9. Epub 2012/10/24. doi: 10.1016/j.taap.2012.10.009 ; PubMed Central PMCID: PMC3508345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wu ZQ, Vos Q, Shen Y, Lees A, Wilson SR, Briles DE, et al. In vivo polysaccharide-specific IgG isotype responses to intact Streptococcus pneumoniae are T cell dependent and require CD40- and B7-ligand interactions. J Immunol. 1999;163(2):659–67. Epub 1999/07/08. . [PubMed] [Google Scholar]
  • 14.Wu ZQ, Khan AQ, Shen Y, Schartman J, Peach R, Lees A, et al. B7 requirements for primary and secondary protein- and polysaccharide-specific Ig isotype responses to Streptococcus pneumoniae. J Immunol. 2000;165(12):6840–8. Epub 2000/12/20. doi: 10.4049/jimmunol.165.12.6840 . [DOI] [PubMed] [Google Scholar]
  • 15.Edwards JR, Prozialeck WC. Cadmium, diabetes and chronic kidney disease. Toxicology and Applied Pharmacology. 2009;238(3):289–93. doi: 10.1016/j.taap.2009.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Han JC, Park SY, Hah BG, Choi GH, Kim YK, Kwon TH, et al. Cadmium induces impaired glucose tolerance in rat by down-regulating GLUT4 expression in adipocytes. Archives of Biochemistry and Biophysics. 2003;413(2):213–20. doi: 10.1016/s0003-9861(03)00120-6 [DOI] [PubMed] [Google Scholar]
  • 17.Lei LJ, Jin TY, Zhou YF. Insulin expression in rats exposed to cadmium. Biomed Environ Sci. 2007;20(4):295–301. Epub 2007/10/24. . [PubMed] [Google Scholar]
  • 18.Xing Y, Xia W, Zhang B, Zhou A, Huang Z, Zhang H, et al. Relation between cadmium exposure and gestational diabetes mellitus. Environ Int. 2018;113:300–5. Epub 2018/01/18. doi: 10.1016/j.envint.2018.01.001 . [DOI] [PubMed] [Google Scholar]
  • 19.Haribhai D, Williams JB, Jia S, Nickerson D, Schmitt EG, Edwards B, et al. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity. 2011;35(1):109–22. Epub 2011/07/05. doi: 10.1016/j.immuni.2011.03.029 [pii] . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jeon EJ, Yoon BY, Lim JY, Oh HJ, Park HS, Park MJ, et al. Adoptive transfer of all-trans-retinal-induced regulatory T cells ameliorates experimental autoimmune arthritis in an interferon-gamma knockout model. Autoimmunity. 2012;45(6):460–9. Epub 2012/05/09. doi: 10.3109/08916934.2012.682666 . [DOI] [PubMed] [Google Scholar]
  • 21.Verhagen J, Wegner A, Wraith DC. Extra-thymically induced T regulatory cell subsets: the optimal target for antigen-specific immunotherapy. Immunology. 2015;145(2):171–81. doi: 10.1111/imm.12458 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sood S, Brownlie RJ, Garcia C, Cowan G, Salmond RJ, Sakaguchi S, et al. Loss of the Protein Tyrosine Phosphatase PTPN22 Reduces Mannan-Induced Autoimmune Arthritis in SKG Mice. J Immunol. 2016. Epub 2016/06/12. doi: 10.4049/jimmunol.1502656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kluger MA, Nosko A, Ramcke T, Goerke B, Meyer MC, Wegscheid C, et al. RORgammat expression in Tregs promotes systemic lupus erythematosus via IL-17 secretion, alteration of Treg phenotype and suppression of Th2 responses. Clinical and experimental immunology. 2017;188(1):63–78. Epub 2016/11/24. doi: 10.1111/cei.12905 ; PubMed Central PMCID: PMC5343349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Holohan DR, Van Gool F, Bluestone JA. Thymically-derived Foxp3+ regulatory T cells are the primary regulators of type 1 diabetes in the non-obese diabetic mouse model. PLOS ONE. 2019;14(10):e0217728. doi: 10.1371/journal.pone.0217728 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ohkura N, Yasumizu Y, Kitagawa Y, Tanaka A, Nakamura Y, Motooka D, et al. Regulatory T Cell-Specific Epigenomic Region Variants Are a Key Determinant of Susceptibility to Common Autoimmune Diseases. Immunity. 2020;52(6):1119–32 e4. Epub 2020/05/05. doi: 10.1016/j.immuni.2020.04.006 . [DOI] [PubMed] [Google Scholar]
  • 26.Wing JB, Tanaka A, Sakaguchi S. Human FOXP3+ Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity. 2019;50(2):302–16. doi: 10.1016/j.immuni.2019.01.020 [DOI] [PubMed] [Google Scholar]
  • 27.Long SA, Buckner JH. CD4+FOXP3+ T regulatory cells in human autoimmunity: more than a numbers game. J Immunol. 2011;187(5):2061–6. Epub 2011/08/23. doi: 10.4049/jimmunol.1003224 ; PubMed Central PMCID: PMC3160735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kitagawa Y, Wing JB, Sakaguchi S. Transcriptional and Epigenetic Control of Regulatory T Cell Development. Prog Mol Biol Transl Sci. 2015;136:1–33. Epub 2015/11/29. doi: 10.1016/bs.pmbts.2015.07.011 . [DOI] [PubMed] [Google Scholar]
  • 29.Sakaguchi S, Takahashi T, Hata H, Nomura T, Sakaguchi N. SKG mice, a new genetic model of rheumatoid arthritis. Arthritis research & therapy. 2003;5(Suppl 3):10–. doi: 10.1186/ar811 . [DOI] [Google Scholar]
  • 30.Sakaguchi S, Sakaguchi N. Animal models of arthritis caused by systemic alteration of the immune system. Current opinion in immunology. 2005;17(6):589–94. Epub 2005/10/11. doi: 10.1016/j.coi.2005.09.017 . [DOI] [PubMed] [Google Scholar]
  • 31.Sakaguchi S, Sakaguchi N, Yoshitomi H, Hata H, Takahashi T, Nomura T. Spontaneous development of autoimmune arthritis due to genetic anomaly of T cell signal transduction: Part 1. Seminars in immunology. 2006;18(4):199–206. Epub 2006/05/23. doi: 10.1016/j.smim.2006.03.007 . [DOI] [PubMed] [Google Scholar]
  • 32.Salmon P. Application of Bone Morphometry and Densitometry by X-Ray Micro-CT to Bone Disease Models and Phenotypes. 2020. p. 49–75. [Google Scholar]
  • 33.Leffel EK, Wolf C, Poklis A, White KL Jr., Drinking water exposure to cadmium, an environmental contaminant, results in the exacerbation of autoimmune disease in the murine model. Toxicology. 2003;188(2–3):233–50. Epub 2003/05/28. doi: 10.1016/s0300-483x(03)00092-1 . [DOI] [PubMed] [Google Scholar]
  • 34.Moudgil KD, Choubey D. Cytokines in autoimmunity: role in induction, regulation, and treatment. J Interferon Cytokine Res. 2011;31(10):695–703. Epub 2011/09/23. doi: 10.1089/jir.2011.0065 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lu J, Liu J, Li L, Lan Y, Liang Y. Cytokines in type 1 diabetes: mechanisms of action and immunotherapeutic targets. Clin Transl Immunology. 2020;9(3):e1122–e. doi: 10.1002/cti2.1122 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Müller R. Guidelines for assessment of bone microstructure in rodents using micro–computed tomography. Journal of Bone and Mineral Research. 2010;25(7):1468–86. doi: 10.1002/jbmr.141 [DOI] [PubMed] [Google Scholar]
  • 37.Donnelly E. Methods for Assessing Bone Quality: A Review. Clinical Orthopaedics and Related Research®. 2011;469(8). doi: 10.1007/s11999-010-1702-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hess EV. Environmental chemicals and autoimmune disease: cause and effect. Toxicology. 2002;181–182:65–70. Epub 2002/12/31. doi: 10.1016/s0300-483x(02)00004-5 . [DOI] [PubMed] [Google Scholar]
  • 39.Buha A, Dukic-Cosic D, Curcic M, Bulat Z, Antonijevic B, Moulis JM, et al. Emerging Links between Cadmium Exposure and Insulin Resistance: Human, Animal, and Cell Study Data. Toxics. 2020;8(3). Epub 2020/09/02. doi: 10.3390/toxics8030063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tan TG, Mathis D, Benoist C. Singular role for T-BET+CXCR3+ regulatory T cells in protection from autoimmune diabetes. Proc Natl Acad Sci U S A. 2016;113(49):14103–8. Epub 2016/11/23. doi: 10.1073/pnas.1616710113 ; PubMed Central PMCID: PMC5150376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.D’Alise AM, Auyeung V, Feuerer M, Nishio J, Fontenot J, Benoist C, et al. The defect in T-cell regulation in NOD mice is an effect on the T-cell effectors. Proc Natl Acad Sci U S A. 2008;105(50):19857–62. Epub 2008/12/17. doi: 10.1073/pnas.0810713105 ; PubMed Central PMCID: PMC2604930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Martin-Orozco N, Chung Y, Chang SH, Wang Y-H, Dong C. Th17 cells promote pancreatic inflammation but only induce diabetes efficiently in lymphopenic hosts after conversion into Th1 cells. European Journal of Immunology. 2009;39(1):216–24. doi: 10.1002/eji.200838475 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Emamaullee JA, Davis J, Merani S, Toso C, Elliott JF, Thiesen A, et al. Inhibition of Th17 cells regulates autoimmune diabetes in NOD mice. Diabetes. 2009;58(6):1302–11. Epub 2009/03/18. doi: 10.2337/db08-1113 ; PubMed Central PMCID: PMC2682686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kuriya G, Uchida T, Akazawa S, Kobayashi M, Nakamura K, Satoh T, et al. Double deficiency in IL-17 and IFN-γ signalling significantly suppresses the development of diabetes in the NOD mouse. Diabetologia. 2013;56(8):1773–80. Epub 2013/05/24. doi: 10.1007/s00125-013-2935-8 . [DOI] [PubMed] [Google Scholar]
  • 45.Serreze DV, Post CM, Chapman HD, Johnson EA, Lu B, Rothman PB. Interferon-gamma receptor signaling is dispensable in the development of autoimmune type 1 diabetes in NOD mice. Diabetes. 2000;49(12):2007–11. doi: 10.2337/diabetes.49.12.2007 [DOI] [PubMed] [Google Scholar]
  • 46.Yi Z, Li L, Garland A, He Q, Wang H, Katz JD, et al. IFN-γ receptor deficiency prevents diabetes induction by diabetogenic CD4+, but not CD8+, T cells. Eur J Immunol. 2012;42(8):2010–8. Epub 2012/08/07. doi: 10.1002/eji.201142374 ; PubMed Central PMCID: PMC3883988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Schurgers E, Billiau A, Matthys P. Collagen-induced arthritis as an animal model for rheumatoid arthritis: focus on interferon-γ. J Interferon Cytokine Res. 2011;31(12):917–26. Epub 2011/09/13. doi: 10.1089/jir.2011.0056 . [DOI] [PubMed] [Google Scholar]
  • 48.Takeuchi Y, Hirota K, Sakaguchi S. Impaired T cell receptor signaling and development of T cell-mediated autoimmune arthritis. Immunol Rev. 2020;294(1):164–76. Epub 2020/01/17. doi: 10.1111/imr.12841 . [DOI] [PubMed] [Google Scholar]
  • 49.Keller KK, Lindgaard LM, Wogensen L, Dagnaes-Hansen F, Thomsen JS, Sakaguchi S, et al. SKG arthritis as a model for evaluating therapies in rheumatoid arthritis with special focus on bone changes. Rheumatology international. 2013;33(5):1127–33. Epub 2012/09/06. doi: 10.1007/s00296-012-2500-7 . [DOI] [PubMed] [Google Scholar]
  • 50.Brama M, Politi L, Santini P, Migliaccio S, Scandurra R. Cadmium-induced apoptosis and necrosis in human osteoblasts: role of caspases and mitogen-activated protein kinases pathways. J Endocrinol Invest. 2012;35(2):198–208. Epub 2011/06/24. doi: 10.3275/7801 . [DOI] [PubMed] [Google Scholar]
  • 51.Chen X, Zhu G, Jin T, Zhou Z, Gu S, Qiu J, et al. Cadmium stimulates the osteoclastic differentiation of RAW264.7 cells in presence of osteoblasts. Biol Trace Elem Res. 2012;146(3):349–53. Epub 2011/11/15. doi: 10.1007/s12011-011-9256-x . [DOI] [PubMed] [Google Scholar]
  • 52.Zhao H, Liu W, Wang Y, Dai N, Gu J, Yuan Y, et al. Cadmium induces apoptosis in primary rat osteoblasts through caspase and mitogen-activated protein kinase pathways. J Vet Sci. 2015;16(3):297–306. Epub 2015/10/02. doi: 10.4142/jvs.2015.16.3.297 ; PubMed Central PMCID: PMC4588015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Boughammoura S, Ben Mimouna S, Chemek M, Ostertag A, Cohen-Solal M, Messaoudi I. Disruption of Bone Zinc Metabolism during Postnatal Development of Rats after Early Life Exposure to Cadmium. Int J Mol Sci. 2020;21(4). Epub 2020/02/16. doi: 10.3390/ijms21041218; PubMed Central PMCID: PMC7072778. [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

David M Lehmann

19 Apr 2021

PONE-D-21-08667

Prenatal cadmium exposure does not induce greater incidence or earlier onset of autoimmunity in the offspring

PLOS ONE

Dear Dr. Barnett,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

All three reviewers make important suggestions on how to improve the manuscript, ranging from content to presentation.  Please go carefully through all review comments below.

Please submit your revised manuscript by Jun 03 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols.

We look forward to receiving your revised manuscript.

Kind regards,

David M. Lehmann, Ph.D.

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

  1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

  1. To comply with PLOS ONE submissions requirements, in your Methods section, please provide additional information on the animal research and ensure you have included details on methods of anesthesia and/or analgesia, and efforts to alleviate suffering.

  1. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

4. PLOS ONE requires that the data underlying each figure and table be made available to the public prior to publication (https://journals.plos.org/plosone/s/data-availability).  Please review the journal’s policy and provide a suitable mechanism to satisfy the requirement. [NOTE: As indicated by the third Reviewer, information related to the flow cytometry algorithmic analysis are also necessary to fulfill this requirement.] 

Additional Editor Comments (if provided):

  • Given the nature of the results (i.e., negative), small sample size, limited statistical power, and apparent lack of study replication, study limitations need to be described in the Discussion section of the manuscript.

Minor concerns -

  • Abstract line 22 - Remove the word "the" from in between "alters" and "immune"

  • Methods, general - Recognizing that systematic review is becoming common practice and that systematic reviews involve an assessment of study quality, I recommend that the authors provide more details in the Methods section.  For example, you should specify the animal husbandry conditions, if the animals were randomized (and how), test material purity, test material source (city, state), and whether, or not all experimental groups were "exposed" concurrently or if the treatments were staggered over time. 

  • Methods, line 189 - Consider adding what the samples were analyzed individually for (i.e., analyzed individually for cytokines (see below).

  • Results, line 249 - Provide a reference, please.

  • Results, line 257 - Change "significant" to "significance"

  • Results, line 270 - 272 - I think it would help the reader if you defined what constitutes a positive result.

  • Figures, general - The text is blurry and difficult to read for several of the figures.  Please adjust the resolution.

  • Figure, legends - I think it would help if you were clear about which mice you were evaluating in the bold text component of each figure legend.  For example, in Figure 1 you specified offspring, but in Figures 2, 3, 4, and 6 the specific animals you are evaluating isn't clearly stated upfront.  The remaining figures are specific to adults and, again, I think it would be better to make that clear upfront.

  • Figure 3 - For consistency with Figures 1, 2, and S1 and ease of reading, please put the figure legend in the top panel.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This manuscript by McCall and colleagues looks at the effect of prenatal cadmium (Cd) exposure on development of autoimmunity in mouse models of type 1 diabetes and arthritis. The takeaway from these studies are that prenatal Cd exposure had little effect on development of specific autoimmune phenotypes in these experimental models.

This is essentially a negative study but also a cautionary tale demonstrating that xenobiotic exposures linked to autoimmunity in both humans and animals (mainly mice) can fail to impact disease development in animal models prone to specific diseases. This is known for other xenobiotics, especially after exposure in adults, and so reporting its occurrence for Cd in prenatal exposure is important.

The authors should provide some comparative discussion on the effects of Cd exposure in adult animals as this is known to elicit autoimmune responses. Interestingly, Cd can induce autoantibodies in healthy mice as well as exacerbate autoimmunity in autoimmune prone strains.

In some situations xenobiotic exposure may not exacerbate or accelerate a specific autoimmune phenotype, but may still elicit a response characteristic of that xenobiotic. As indicated above, Cd can induce and/or exacerbate anti-nuclear antibodies (ANA). Thus one aspect worth considering is whether the NOD and SKG mice had increases in ANA following prenatal Cd exposure. This would help establish whether this Cd induced response in adult mice also occurs following prenatal exposure in different models of autoimmunity.

Minor points

1) The text within the figures lacks adequate resolution.

2) The x axis in Figure 2 needs text to identify the groups.

3) In Figure 3 there is no indication of any differences in TNF-a, but the control/non-diabetic mean is much greater than the other groups. This stands in contrast to the IL-17F values where seemingly smaller differences are statistically different.

4) The changes described in the micro-CT images in Figure 5 should be indicated by arrows or circles.

Reviewer #2: Based on previously results, authors investigated if Cd exposure at environmental level could can favor the onset and severity of autoimmune diseases in two animal models. The effect of prenatal Cd (10 ppm) on the development of diabetes (NOD mice) and arthritis (zymosan-induced autoimmune arthritis in SKG mice) were investigated. Small differences were observed in exposed animals compared to control mice, that were judged not biological relevant, leading to the conclusion that prenatal exposure to Cd did not increase the propensity to develop autoimmune diseases.

Below some concerns:

- In the introduction line 78, the nature of the persistent changes in thymus and spleen should be better defined.

- In some group the number of animal is very low (n=4), how can this affect the results? why only 4 animals? taking into account the biological variability in the response, has a statistical analysis been made to define the optimal experimental design to highlight differences and establish the appropriate number of animals?

- Considering that Cd is an environmental contaminant, why exposure did not continue after weaning? In real life, we can imagine continued exposure.

- Figure 1B. In the legend it is not reported at what time point the percent diabetic is referred to. Likely 18-22 weeks, but this should be specified. Interesting is the gender difference, with females exposed to Cd showing a delay in the onset. In this model, if we want to see some effect of cadmium, at most we can hypothesize a protective effect.

- Figure 2. How can authors explain the lack of decrease in Treg previously observed? How can this be transposed to humans? Are the percentage the same as well?

- Figure 4. In this model, it seems an earlier onset in males exposed to Cd.

- Where the levels of anti-inflammatory cytokines affected?

- In the results sections, the nature of changes must be specified.

- In the Discussion, repetition of the results should be avoided.

- Line 402, what do the authors mean with ‘disrupt’?

Reviewer #3: There is as major inconsistency between the text and the data for cytokine production. In Figure 2, the levels of IL-17A and IL-17F show no obvious changes with Cd treatment, whereas TNFalpha levels in male mice are approximately ten-fold lower in non-diabetic mice after Cd treatment. The significance values are shown for two comparisons for IL-17F, but the much larger TNF difference is ignored. This mismatch between the text and the data needs to be resolved.

Publication of negative data is important to provide other investigators with planning information, and to reduce duplication of results. However, in contrast to positive results, it is difficult to identify a statistical significance for a lack of effect. Thus the number of replicates, and the consistency of the results, become more important when reporting negative results. This study used relatively small groups of mice, and there is no indication of whether the experiments were repeated.

In a previous study (ref. 11) on prenatal exposure of mice to Cd, this group found an increase in antibody response and IFNg production, and a decrease in nTreg cells, using two or three experiments with 5-8 mice per condition each. However, in the current study in the NOD mouse, a significant decrease in nTreg cells was not observed. This could have been due to differences between the mouse strains, but it could also be related to the small numbers of mice in the current study. Specifically, in Figure 5 of ref 11, there is a decrease of nTreg cells from about 1.65% to 1.3% with Cd-treated females (males changed in the opposite direction). In the current study, the mean differences were larger, i.e. in Fig. 2, females showed about 3% and 2.1% nTregs with and without Cd. Thus the averages in the two studies are concordant for females (not males) but the authors draw different conclusions because of the increased spread of the data in the current study. These results are a good example of the way that a lack of significant effect may be due to low sample numbers. Phrased differently, the current study does not appear to have been powered sufficiently to detect changes at the level shown in the previous study.

Given that in their previous study, the authors found an impressive difference in IFNg levels after prenatal cadmium exposure, why was IFNg not measured in this study?

Demonstration of the detailed flow cytometry algorithmic analysis (or manual gating) that was used to enumerate cell populations should be provided as supplementary data. This is important for any flow cytometry data, but particularly for Treg cells that have less well-defined marker staining compared to other T cell markers.

At the end of the introduction, the authors state that “unfortunately” Cd did not increase automimmunity in their model. While it is reasonable that the authors are disappointed in the outcome of their hypothesis, at face value their model may suggest that Cd does not have a strong in utero effect on autoimmune disease, which could be good news?

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: K. Michael Pollard

Reviewer #2: No

Reviewer #3: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 Sep 3;16(9):e0249442. doi: 10.1371/journal.pone.0249442.r002

Author response to Decision Letter 0


3 Aug 2021

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming.

File naming and author affiliations have been updated

2. To comply with PLOS ONE submissions requirements, in your Methods section, please provide additional information on the animal research and ensure you have included details on methods of anesthesia and/or analgesia, and efforts to alleviate suffering.

Additional information was included.

3. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

All figures are included in the manuscript or uploaded to Figshare and cited.

4. PLOS ONE requires that the data underlying each figure and table be made available to the public prior to publication

All raw data used to generate graphs presented as means are included in new table “S1 Table.” Flow cytometry gating strategies are found in “S1 Fig.”

Additional Editor Comments (if provided):

• Given the nature of the results (i.e., negative), small sample size, limited statistical power, and apparent lack of study replication, study limitations need to be described in the Discussion section of the manuscript.

Added samples to the bone microarchitecture experiments. Text has been updated to clarify the replicates and study limitations have been added to the discussion.

Minor concerns -

• Abstract line 22 - Remove the word "the" from in between "alters" and "immune" Fixed

• Methods, general - Recognizing that systematic review is becoming common practice and that systematic reviews involve an assessment of study quality, I recommend that the authors provide more details in the Methods section. For example, you should specify the animal husbandry conditions, if the animals were randomized (and how), test material purity, test material source (city, state), and whether, or not all experimental groups were "exposed" concurrently or if the treatments were staggered over time. Added details

• Methods, line 189 - Consider adding what the samples were analyzed individually for (i.e., analyzed individually for cytokines (see below). Added text to clarify

• Results, line 249 - Provide a reference, please. Added

• Results, line 257 - Change "significant" to "significance" Fixed

• Results, line 270 - 272 - I think it would help the reader if you defined what constitutes a positive result. Additional text was added to clarify

• Figures, general - The text is blurry and difficult to read for several of the figures. Please adjust the resolution. Use PACE to upload figures.

• Figure, legends - I think it would help if you were clear about which mice you were evaluating in the bold text component of each figure legend. For example, in Figure 1 you specified offspring, but in Figures 2, 3, 4, and 6 the specific animals you are evaluating isn't clearly stated upfront. The remaining figures are specific to adults and, again, I think it would be better to make that clear upfront. Legends were updated to clarify animals analyzed

• Figure 3 - For consistency with Figures 1, 2, and S1 and ease of reading, please put the figure legend in the top panel. Changed

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

________________________________________

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

________________________________________

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

________________________________________

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

________________________________________

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: This manuscript by McCall and colleagues looks at the effect of prenatal cadmium (Cd) exposure on development of autoimmunity in mouse models of type 1 diabetes and arthritis. The takeaway from these studies are that prenatal Cd exposure had little effect on development of specific autoimmune phenotypes in these experimental models.

This is essentially a negative study but also a cautionary tale demonstrating that xenobiotic exposures linked to autoimmunity in both humans and animals (mainly mice) can fail to impact disease development in animal models prone to specific diseases. This is known for other xenobiotics, especially after exposure in adults, and so reporting its occurrence for Cd in prenatal exposure is important.

The authors should provide some comparative discussion on the effects of Cd exposure in adult animals as this is known to elicit autoimmune responses. Interestingly, Cd can induce autoantibodies in healthy mice as well as exacerbate autoimmunity in autoimmune prone strains.

In some situations xenobiotic exposure may not exacerbate or accelerate a specific autoimmune phenotype, but may still elicit a response characteristic of that xenobiotic. As indicated above, Cd can induce and/or exacerbate anti-nuclear antibodies (ANA). Thus one aspect worth considering is whether the NOD and SKG mice had increases in ANA following prenatal Cd exposure. This would help establish whether this Cd induced response in adult mice also occurs following prenatal exposure in different models of autoimmunity. Performed ANA screen in SKG sera.

Minor points

1) The text within the figures lacks adequate resolution. Upload using PACE

2) The x axis in Figure 2 needs text to identify the groups. Removed ticks on the x-axis so they didn’t distract from the legend.

3) In Figure 3 there is no indication of any differences in TNF-a, but the control/non-diabetic mean is much greater than the other groups. This stands in contrast to the IL-17F values where seemingly smaller differences are statistically different. The data originally in the graph were incorrect. The stats were performed on the raw data. The graph has been updated with the correct data.

4) The changes described in the micro-CT images in Figure 5 should be indicated by arrows or circles. Added

Reviewer #2: Based on previously results, authors investigated if Cd exposure at environmental level could can favor the onset and severity of autoimmune diseases in two animal models. The effect of prenatal Cd (10 ppm) on the development of diabetes (NOD mice) and arthritis (zymosan-induced autoimmune arthritis in SKG mice) were investigated. Small differences were observed in exposed animals compared to control mice, that were judged not biological relevant, leading to the conclusion that prenatal exposure to Cd did not increase the propensity to develop autoimmune diseases.

Below some concerns:

- In the introduction line 78, the nature of the persistent changes in thymus and spleen should be better defined. Clarified reduction in nTregs in text.

- In some group the number of animal is very low (n=4), how can this affect the results? why only 4 animals? taking into account the biological variability in the response, has a statistical analysis been made to define the optimal experimental design to highlight differences and establish the appropriate number of animals? Two mice from each group were added to the analysis. microCT data was performed on a representative subset of animals that were scored using the arthritic index method. Both methods showed no difference in disease severity.

- Considering that Cd is an environmental contaminant, why exposure did not continue after weaning? In real life, we can imagine continued exposure. This is something that can be addressed in the future; however, for this study we wanted to specifically test the in utero exposure.

- Figure 1B. In the legend it is not reported at what time point the percent diabetic is referred to. Likely 18-22 weeks, but this should be specified. Interesting is the gender difference, with females exposed to Cd showing a delay in the onset. In this model, if we want to see some effect of cadmium, at most we can hypothesize a protective effect. Added discussion regarding “protection” in the discussion.

- Figure 2. How can authors explain the lack of decrease in Treg previously observed? How can this be transposed to humans? Are the percentage the same as well? Clarified that these are different mouse models.

- Figure 4. In this model, it seems an earlier onset in males exposed to Cd. This is consistent with the published data of the model.

- Where the levels of anti-inflammatory cytokines affected? Unfortunately, these were not measured.

- In the results sections, the nature of changes must be specified. Changed ambiguous “differences” to reduced or increased.

- In the Discussion, repetition of the results should be avoided. Moved specific data and p values to result.

- Line 402, what do the authors mean with ‘disrupt’? clarified

Reviewer #3: There is as major inconsistency between the text and the data for cytokine production. In Figure 2, the levels of IL-17A and IL-17F show no obvious changes with Cd treatment, whereas TNFalpha levels in male mice are approximately ten-fold lower in non-diabetic mice after Cd treatment. The significance values are shown for two comparisons for IL-17F, but the much larger TNF difference is ignored. This mismatch between the text and the data needs to be resolved. The data originally in the graph were incorrect. The stats were performed on the raw data. The graph has been updated with the correct data.

Publication of negative data is important to provide other investigators with planning information, and to reduce duplication of results. However, in contrast to positive results, it is difficult to identify a statistical significance for a lack of effect. Thus the number of replicates, and the consistency of the results, become more important when reporting negative results. This study used relatively small groups of mice, and there is no indication of whether the experiments were repeated. Clarified repeats in methods.

In a previous study (ref. 11) on prenatal exposure of mice to Cd, this group found an increase in antibody response and IFNg production, and a decrease in nTreg cells, using two or three experiments with 5-8 mice per condition each. However, in the current study in the NOD mouse, a significant decrease in nTreg cells was not observed. This could have been due to differences between the mouse strains, but it could also be related to the small numbers of mice in the current study. Specifically, in Figure 5 of ref 11, there is a decrease of nTreg cells from about 1.65% to 1.3% with Cd-treated females (males changed in the opposite direction). In the current study, the mean differences were larger, i.e. in Fig. 2, females showed about 3% and 2.1% nTregs with and without Cd. Thus the averages in the two studies are concordant for females (not males) but the authors draw different conclusions because of the increased spread of the data in the current study. These results are a good example of the way that a lack of significant effect may be due to low sample numbers. Phrased differently, the current study does not appear to have been powered sufficiently to detect changes at the level shown in the previous study.

Included figure without dividing the mice by disease state to show that Cd itself is not inducing Treg changes in the model. Increased animal numbers where possible and added additional assays.

Given that in their previous study, the authors found an impressive difference in IFNg levels after prenatal cadmium exposure, why was IFNg not measured in this study? Added paragraph in discussion

Demonstration of the detailed flow cytometry algorithmic analysis (or manual gating) that was used to enumerate cell populations should be provided as supplementary data. This is important for any flow cytometry data, but particularly for Treg cells that have less well-defined marker staining compared to other T cell markers. Added gating strategy in supplemental figure 1.

At the end of the introduction, the authors state that “unfortunately” Cd did not increase automimmunity in their model. While it is reasonable that the authors are disappointed in the outcome of their hypothesis, at face value their model may suggest that Cd does not have a strong in utero effect on autoimmune disease, which could be good news? Updated introduction to include the positive aspect of the data.

________________________________________

Attachment

Submitted filename: Response to Reveiwers.docx

Decision Letter 1

David M Lehmann

24 Aug 2021

Prenatal cadmium exposure does not induce greater incidence or earlier onset of autoimmunity in the offspring

PONE-D-21-08667R1

Dear Dr. Barnett,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

David M. Lehmann, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: (No Response)

Reviewer #2: I thank the Authors. I have no further comments, as Authors have properly addressed all my concerns.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: Kenneth Michael Pollard

Reviewer #2: No

Acceptance letter

David M Lehmann

26 Aug 2021

PONE-D-21-08667R1

Prenatal cadmium exposure does not induce greater incidence or earlier onset of autoimmunity in the offspring

Dear Dr. Barnett:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. David M. Lehmann

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Gating strategies for flow cytometry panels.

    (TIF)

    S2 Fig. Spleen cell phenotypes of cadmium exposed NOD mice.

    Dam and sire NOD mice were exposed to cadmium as described. At 18 weeks (females) and 22 weeks (males), the spleens were ablated and processed for flow cytometric analysis. CD4+, CD8+ and CD19+ were quantified. Symbols represent the data for individual mice and the black bar denotes the mean. Males N = 6–16; Females N = 13–19.

    (TIF)

    S3 Fig. Spleen cell cytokine production by cadmium exposed NOD mice.

    Dam and sire NOD mice were exposed to cadmium as described. At 18 weeks (females) and 22 weeks (males), the spleens of the offspring were ablated and stimulated with anti-CD3+anti-CD28 for 72h. Cytokine levels were measured using MSD multiplex plates. No differences were noted in production of these cytokines between any group. Symbols represent the data for individual mice and the black bar denotes the median. Males N = 5–17; Females N = 4–14.

    (TIF)

    S4 Fig. Prenatal Cd exposure does not induce ANA in SKG model.

    Serum autoantibodies were measured by ANA Hep Screen ELISA.

    (TIF)

    S5 Fig. SKG mice body weight at time of induction and AI of left rear paws.

    A) Body weights of male and female mice at the time of arthritis induction with zymosan A. Symbols represent the data for individual mice and the black bar denotes the median. B) Left rear paw arthritic indices. No differences were noted between control and cadmium exposure within each sex. Data are shown as mean ± SEM.

    (TIF)

    S6 Fig. BMD analysis of the femurs of arthritic Control and Cd-exposed SKG offspring.

    Bone mineral density (BMD) was measured at the distal femur using the same Volume of Interest (1.05 mm) following calibration with calcium hydroxyapatite phantoms of known density: 0.25 and 0.75 g/cm3. Phantoms were prepared, scanned, and reconstructed with the same parameters as Fig 7. Data are shown as mean ± SD.

    (TIF)

    S1 Table. Raw data used for graphs presented as means.

    (XLSX)

    Attachment

    Submitted filename: Response to Reveiwers.docx

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES