Abstract
The gut microbiota plays an important role throughout the lifespan in maintaining host health, and several factors can modulate microbiota composition including diet, exercise, and environmental exposures. Maternal microbiota is transferred to offspring during early life; thus, environmental exposures before gestation may also modulate offspring microbiota. Here we aimed to investigate the effects of maternal exposure to dioxin-like polychlorinated biphenyls (PCBs) on the microbiota of aged offspring and to determine if lifestyle factors, including maternal exercise or offspring high-fat feeding alter these associations. To test this, dams were exposed to PCB 126 (0.5 μmole/kg body weight) or vehicle oil by oral gavage during preconception, gestation, and during lactation. Half of each group was allowed access to running wheels for ≥7 days before and during pregnancy and up through day 14 of lactation. Female offspring born from the 4 maternal groups (PCB exposure or not, with/without exercise) were subsequently placed either on regular diet or switched to a high-fat diet during adulthood. Microbiota composition was quantified in female offspring at 49 weeks of age by 16S rRNA sequencing. Maternal exposure to PCB 126 resulted in significantly reduced richness and diversity in offspring microbiota regardless of diet or exercise. Overall compositional differences were largely driven by offspring diet, but alterations in specific taxa due to maternal PCB 126 exposure, included the depletion of Verrucomicrobiaceae and Akkermansia muciniphila, and an increase in Anaeroplasma. Perturbation of microbiota due to PCB 126 may predispose offspring to a variety of chronic diseases later in adulthood.
Keywords: Polychlorinated biphenyls, gut microbiota, DoHaD, Maternal Exercise, persistent organic pollutants, mice, pregnancy
Introduction
Gut microbiota refers to the set of microorganisms such as bacteria, viruses, protozoa, and fungi that coexist in the digestive tract and make up an integral part of the body [1]. Numerous factors impact the form and function of gut microbiota, with dysbiosis and modulation of microbiota impacting human health [2]. A host’s environment and lifestyle have long been shown to modulate microbiota taxa and diversity with much emphasis focusing on diet and personal exposures including cigarette smoking. Recently, exposures to ubiquitous environmental chemicals including persistent organic pollutants such as polychlorinated biphenyls (PCBs) have also been shown to directly or indirectly impact gut health [3-7]. Although PCBs are no longer manufactured in the United States since they were banned in 1979 due to adverse health effects on multiple organ systems [8-11], they continue to contaminate our environment through multiple routes including improper disposal or incineration of contaminated waste and leaks from old transformers and capacitors[12, 13]. Currently, the major source of human exposure to PCBs is through consumption of contaminated food especially fish from contaminated lakes and rivers, meat, and dairy products [12].
Microbiota is vertically transferred from mothers to offspring, thus exposures that impact the microbiota of mothers prior to or during conception—as well as during pregnancy—may also impact their offspring’s microbiota [14, 15]. As modulation of normal microbiota homeostasis is associated with many chronic diseases, this paradigm can be seen as a growing research area related to the Developmental Origins of Health and Disease (DOHaD) hypothesis [16, 17]. Studies in adult rodents have shown that PCBs and dioxin-like pollutant mixtures can significantly impact gut microbiota diversity and taxa [3, 7, 18-20]. We and others have shown that PCB exposure significantly decreases the relative abundance of Proteobacteria, Bacteroidales family S7-25, and Alistipes compared with unexposed controls [6]. A decrease in diversity and an increase in the Firmicutes to Bacteroidetes ratio (F/B ratio) has also been demonstrated [2, 21]. These dioxin-like pollutants have been linked to metabolic dysfunction, reproductive disorders, neurobehavioral impairments, and immune dysfunction [22], which is potentially mediated by alterations to microbiota.
Several studies have implicated maternal diet and exercise with modulation of offspring gut microbiota [19, 23-27], but to our knowledge, no group has focused on other environmental exposures such as dioxin-like pollutants. In addition, only a few studies in adult rodents have investigated if other factors known to impact the microbiota, such as diet and exercise, may mask or modulate toxicant-induced changes in the gut microbiota [6, 18]. Our present study seeks to examine the impact of maternal exposure to the model dioxin-like pollutant, PCB 126, on offspring gut microbiota, and if maternal exercise or offspring diet can mask or exacerbate these changes. We hypothesized that perinatal exposure to PCBs will result in decreased alpha and compositional differences in the gut microbiota of the offspring, despite the offspring never being directly exposed to PCBs via oral gavage. We further hypothesized that these changes in offspring may be attenuated by maternal exercise, and that feeding offspring with a high-fat diet will exacerbate the effects on the gut microbiota.
Materials and Methods
In vivo study design
These studies were carried out at the University of Kentucky according to an approved Institutional Animal Care and Use Committee (IACUC # 2009-0503) protocol. Two to four-month-old proven ICR breeders from Envigo Harlan were shipped to the University of Kentucky and housed four per cage for one week to acclimate. During this period, they were on regular mouse diet 2018 (Harlan). The following week, they were individually housed in circadian lightbox cabinets (Phenome Technologies, Lincolnshire, IL) for habituation and placed on maternal breeder diet 5008 (Lab Diet, St. Louis, MO). A week after being in the light box, dams were weighed and placed in the following groups: Group 1- Sedentary/Vehicle (Sed/Veh) exposed to safflower oil with no access to a running wheel, Group 2- Exercise/Vehicle (Ex/Veh) exposed to safflower oil with voluntary access to a running wheel, Group 3-Sedentary/PCB (Sed/PCB) exposed to PCB126 (0.5 μmole/kg body weight PCB126) with no access to a running wheel, and Group 4- Exercise/PCB exposed to PCB126 with voluntary access to a running wheel. All mice were housed in an environmentally controlled vivarium between 20-22°C with unlimited access to food and water under a controlled photoperiod (14:10-h light-dark). If applicable, mice had open access to the running wheels that were mounted within each cage (Lincolnshire, IL). A mechanical counter was used to record wheel rotations to a desktop computer via ClockLab software (Actimetrics, Wilmette, IL). Sedentary female mice were housed in nearly identical cages that did not contain running wheels as we did in our previous study[28]. Dams were allowed to exercise ≥7 days before and during pregnancy and up through day 14 of lactation. The exercise was completely voluntary.
Dams were exposed 3 times to PCB 126: The first exposure was administered 72 hours before mating. The second exposure was administered on approximate gestational day 7 and the last exposure was administered on a postnatal day 7 (PND7) during lactation. The current study utilizes a similar dosing paradigm in previous studies [29, 30]. The pregnant dams in the exercise cohort had continual access to running wheels throughout pregnancy until 14 days after giving birth, at which point wheels were removed to prevent possible harm to growing pups Litters were culled to eight or nine pups ~48 h after birth. Pups were cross-fostered from other litters from the same group if they did not have at least eight pups.
Female offspring were weaned at 21d (3 weeks) and placed on Teklad Global 18% Protein Rodent Diet No. 2018 (regular mouse diet 2018). The housing density was five per cage and each weanling cage had a (mix of litter)-one litter from a different dam within the same treatment group. At 39 weeks of age, half of the female offspring were placed on high-fat feeding D12492 with 60 kcal % fat (Research Diets; New Brunswick, NJ) as a means to induce a secondary metabolic hit in adulthood in an attempt to further elucidate PCB 126 induced impacts. The other half of the offspring remained on regular diet for the duration of the study. The offspring themselves did not have access to running wheels during any portion of the study. At the end of the study, offspring were fasted for 3 hours before euthanasia via carbon dioxide, and cecum samples were collected at 49 weeks of age, snap-frozen in liquid nitrogen, and stored at −80 degrees C until analysis (offspring n=5 from each group). As is typical of DOHaD studies, offspring sample size indicates the number of dams/litters (only one offspring per litter were included in each of the 4 maternal exposure/exercise groups; though siblings were included in the offspring dietary feeding portion of the experiment). Male offspring were used for different experiments, so were not included in the microbiota studies.
DNA extraction and 16S rRNA sequencing
DNA extraction and 16S rRNA gene sequencing of the V4-V5 region was conducted by the Environmental Sample Preparation and Sequencing Facility (ESPSF) at Argonne National Laboratory as discussed previously[2]. Data processing was conducted using Quantitative Insights into Microbial Ecology (QIIME version 1.9) by ESPSF. Reads were joined using the command join_paired_ends.py and demultiplexed using the command “split_libaries_fastq.py” with the default parameters of 1.5 mismatches. To pick de novo operational taxonomic units (OTUs), the command “pick_de_novo_otus.py” was used and observations that were present less than twice were filtered from the OTU table before further analysis. Read depths in the filtered, unrarefied table ranged from 56 to 174,324 (Median [Q1; Q3]=85,836 [72,674; 102,775]). The sample with the minimum depth of 56 was considered defective (2nd smallest depth=56,285) and was excluded from analyses. A rarefied OTU table was calculated for alpha and beta diversity analyses, which was derived using a multiple rarefaction algorithm [31] at a depth of 56,285 (retaining all non-defective samples). The unrarefied OTU table was using for taxonomic testing, described in detail below.
Statistical analyses
A power calculation was performed to determine the detectable effect size for the association between alpha diversity and PCB exposure by diet/exercise using two-way ANOVA with interaction terms. Assuming a type I error rate of 5%, we are able to detect an effect size of (quantified using Cohen’s f) of 0.46 with 80% power. According to Cohen [32], 0.10 is considered a small effect size, 0.30 is considered a medium effect size, and 0.50 is considered a large effect size. Therefore, we are only able to detect interactions with large effect sizes given our current sample size.
Alpha diversity metrics (richness, Pielou’s evenness, and Faith’s phylogenetic diversity) were calculated using the R packages picante [33] and vegan[34]. Unweighted and weighted UniFrac beta diversity metrics were calculated using the R package phyloseq [35] Weighted Unifrac accounts for the abundance of the observed organisms, whereas unweighted Unifrac only considers the presence or absence of organisms [36]. Differences in alpha diversity metrics were tested using linear regression models, first examining the 3-way interaction (PCB*exercise*diet), followed by 2-way interactions and main effects. Compositional difference testing was performed with PERMANOVA using the R package vegan, testing 3-way and 2-way interactions, and main effects. Principal coordinate analysis (PCoA) was utilized to visually examine compositional distinction across experimental conditions. While examining compositional shifts in the microbiota is helpful to observe broad changes, assessing specific taxonomic differences, specifically at the family, genus, and OTU level, allows for the assessment of individual groups of microbes impacted and provides a basis for an inference of microbial functionality. The R package DESeq2 [37] was used for taxonomic testing. Multiple testing correction was performed using Benjamini and Hochberg false discovery rate (FDR) correction with pFDR<0.05 considered significant. For the dam and terminal body weight measurements, and F/B ratio, the statistical significance for all (p<0.05) was determined by Two-way or Three-way ANOVA analysis and posthoc comparisons by the Holm-Sidak method.
Results
Anthropometrics
This dose of PCB 126 or exercise paradigm did not impact body weights of the dams in any group during the duration of the study (Supplemental Figure 1). Neither maternal exercise nor PCB exposure significantly affected offspring body weight, but high-fat-fed offspring weighed significantly more than regular diet -fed mice (p=0.009) (Supplemental Figure 2). PCB exposure did not impact on running distance of dams for any group (data not shown).
Gut microbiota differences in alpha diversity by PCB exposure, diet, and exercise
When alpha diversity was assessed, no significant 3-way or 2-way interactions were noted for all three metrics (Table 1). After adjusting for both diet and exercise, significant main effects were observed for PCB 126 exposure for both richness (p=0.045) and Faith’s diversity (p=0.039), in which PCB exposure resulted in lower levels of both metrics. Additionally, significant main effects of high-fat diet consumption were observed for richness (p<0.001), evenness (p<0.001), and diversity (p<0.001), in which high-fat diet consumption yielded lower levels compared to the regular fat diet. No significant effects of maternal exercise were observed.
Table 1:
Differences in alpha diversity by PCB exposure, diet, and exercise.
| Richness | Pielou’s Evenness | Faith’s Diversity | |
|---|---|---|---|
| β (95% CI); p-value | |||
| 3-Way Interactions a | |||
| PCB*Exercise*Diet | −69 (−646, 508); p=0.81 | −0.03 (−0.12, 0.06); p=0.48 | −17.2 (−50.1, 15.8); p=0.30 | 
| 2-Way Interactions b | |||
| PCB* Exercise | −18 (−302, 265); p=0.90 | 0.02 (−0.03, 0.06); p=0.46 | 1.6 (−14.8, 18.1); p=0.84 | 
| PCB*Diet | 141 (−143, 425); p=0.32 | 0.02 (−0.02, 0.06); p=0.33 | 4.0 (−12.4, 20.5); p=0.62 | 
| Exercise*Diet | −167 (−451, 116); p=0.24 | 0.001 (−0.04, 0.04); p=0.98 | −13.1 (−29.5, 3.4); p=0.12 | 
| Main Effects c | |||
| PCB (Exposed vs. Unexposed) | −144 (−284, −3); p=0.045 | −0.01 (−0.03, 0.01); p=0.28 | −8.6 (−16.8, −0.5); p=0.039 | 
| Exercise (Yes vs. No) | 33 (−108, 173); p=0.64 | 0.01 (−0.01, 0.03); p=0.40 | 2.4 (−5.8, 10.6); p=0.56 | 
| Diet (High Fat vs. Regular) | −325 (−465, −185); p<0.001 | −0.07 (−0.09, −0.04); p<0.001 | −28.2 (−36.4, −20.0); p<0.001 | 
In a linear model including all main effects, 2-way interactions, and the 3-way interaction
In a linear model including all main effects and 2-way interactions
In a linear model including all main effects
Gut microbiota compositional differences by PCB exposure, diet, and exercise
When compositional differences were visually assessed, a large effect of diet most clearly separated microbiota compositions (Supplemental Figure 3 and Supplemental Figure 4). When associations were formally tested in PERMANOVA models (Table 2), the 3-way interaction for Weighted UniFrac distance had a p-value of 0.056; thus, exploratory sub-group analyses was completed (Table 3). Similar significant interactions between PCB exposure and exercise were observed for mice on a high-fat diet (R2=0.098; p=0.042) and a regular diet (R2=0.094; p=0.030). In weighted UniFrac subgroup analyses, a PCB exposure effect was only significant among offspring who were from exercised mothers and fed a regular diet (R2=0.175; p=0.033). Similar results were seen for Unweighted UniFrac distances (Tables 2 and 3). If interaction effects are ignored, significant main effects were observed for diet only (Table 2), using both unweighted and weighted UniFrac (diet explains 13% and 32% of the variability in unweighted and weighted UniFrac distances, respectively (both p<0.001)).
Table 2:
Compositional differences by PCB exposure, diet, and exercise.
| Unweighted UniFrac | Weighted UniFrac | |||
|---|---|---|---|---|
| R2 | p-value | R2 | p-value | |
| 3-Way Interaction a | ||||
| PCB*Exercise*Diet | 0.028 | 0.092 | 0.034 | 0.056 | 
| 2-Way Interactions b | ||||
| Exercise*Diet | 0.028 | 0.091 | 0.020 | 0.26 | 
| Diet*PCB | 0.025 | 0.18 | 0.017 | 0.39 | 
| Exercise*PCB | 0.028 | 0.084 | 0.030 | 0.090 | 
| Main Effects c | ||||
| PCB | 0.027 | 0.14 | 0.021 | 0.26 | 
| Diet | 0.133 | <0.001 | 0.321 | <0.001 | 
| Exercise | 0.027 | 0.12 | 0.033 | 0.072 | 
In a PERMANOVA model including all main effects, 2-way interactions, and the 3-way interaction
In a PERMANOVA model including all main effects and 2-way interactions
In a PERMANOVA model including all main effects
Table 3:
Compositional differences by PCB exposure – subgroup specific effects.
| Unweighted UniFrac | Weighted UniFrac | |||
|---|---|---|---|---|
| R2 | p-value | R2 | p-value | |
| 2-Way Interactions within Subgroups | ||||
| PCB*Diet among Exercised | 0.056 | 0.094 | 0.067 | 0.069 | 
| PCB*Diet among Non-Exercised | 0.050 | 0.21 | 0.039 | 0.29 | 
| PCB*Exercise among High Fat Diet | 0.062 | 0.040 | 0.098 | 0.042 | 
| PCB*Exercise among Regular Diet | 0.068 | <0.001 | 0.094 | 0.030 | 
| PCB Effect within Subgroups | ||||
| PCB among Exercised, High Fat Diet | 0.138 | 0.061 | 0.189 | 0.077 | 
| PCB among Exercised, Regular Diet | 0.144 | 0.009 | 0.175 | 0.033 | 
| PCB among Non-Exercised, High Fat Diet | 0.116 | 0.23 | 0.175 | 0.11 | 
| PCB among Non-Exercised, Regular Diet | 0.134 | 0.016 | 0.131 | 0.25 | 
Examination of the F/B ratio was employed to assess any interactions in broad compositional changes, as Firmicutes and Bacteroidetes are the two predominant phyla in the mammalian gut microbiota (Supplemental Figure 5). No significant 3-way or 2-way interactions were noted for the F/B ratio. However, a significant main effect of exercise was noted, with a lower F/B ratio observed in the offspring of exercised mothers (p=0.034). Additionally, a significant main effect of diet was noted, with a higher F/B ratio observed in high-fat-fed mice (p=0.015).
Taxonomic Differences Associated with PCB Exposure
For family-level abundances, after adjusting for diet and exercise, PCB exposure resulted in a significant increase in Anaeroplasmataceae abundance (log2 fold change=3.07; pFDR=0.009), as well as a significant reduction in Verrucomicrobiaceae abundance (log2 fold change=−2.69; pFDR<0.001), after adjusting for diet and exercise (Figure 1). At the genus level, PCB exposure resulted in a significant reduction in Akkermansia abundance compared to the vehicle-treated group (log2FC=−2.78; pFDR<0.001), as well as a significant increase in Anaeroplasma (log2FC=2.98; pFDR=0.014) and Prevotella (log2FC=2.84; pFDR=0.033) (Figure 2). At the OTU level, a total of 15 OTUs were significantly associated with PCB exposure after diet and exercise adjustment, the majority of which (12/15) were associated with decreased abundance following PCB exposure (Figure 3). Specifically, PCB exposure significantly decreased the abundance of OTUs in the families Alcaligenaceae (Sutterella sp.), Desulfovibrionaceae, Erysipelotrichaceae, Ruminococcaceae, and Verrucomicrobiaceae (Akkermansia muciniphila), while it significantly increased the abundance of an OTU in the family Anaeroplasmataceae (Anaeroplasma sp.)
Figure 1:
Families significantly associated with PCB exposure (pFDR<0.05), after adjusting for diet and exercise. Log2 fold change estimates compare PCB exposed to unexposed (positive: higher abundance in PCB exposed; negative: lower abundance in PCB exposed).
Figure 2:
Genera significantly associated with PCB exposure (pFDR<0.05), after adjusting for diet and exercise. Log2 fold change estimates compare PCB exposed to unexposed (positive: higher abundance in PCB exposed; negative: lower abundance in PCB exposed).
Figure 3:
OTUs significantly associated with PCB exposure (pFDR<0.05), after adjusting for diet and exercise. Log2 fold change estimates compare PCB exposed to unexposed (positive: higher abundance in PCB exposed; negative: lower abundance in PCB exposed).
Discussion
The gut microbiota is a sensitive marker of lifestyle factors including diet and exercise but also may be a sensitive marker of environmental pollutant exposure [38, 39]. Each of these factors during the perinatal period has been demonstrated to be involved in shaping the long-term health and disease risk of the offspring. Additionally, the perinatal period is important in shaping host microbial composition, which is believed to be a component contributing to offspring health and chronic disease risk [40, 41]. Thus, in the present study, we sought to examine the impact of maternal toxicant exposure on offspring gut microbial composition and to determine if other factors known to impact gut health, specifically diet and exercise, may modulate PCB-induced changes. Our results showed that maternal exposure to PCB 126 resulted in significantly reduced alpha diversity in offspring gut microbiota at during adulthood and changes in specific taxa, but significant interactions with diet or exercise were not observed.
Exposure to PCBs is commonly through dietary means thus PCB exposure could detrimentally impact the gut microbial composition and the gastrointestinal environment. A previous 14-week study, that used similar PCB 126 concentrations, also found that PCB exposure resulted in a reduction in alpha diversity measures [2], but this was completed in non-pregnant mice. It was observed in this study that PCB exposure induces shifts in specific microbial populations including lower levels of Clostridiales, Ruminococci, and Lactobacilli, and increased levels of Akkermansia [2]. These pollutant-induced microbial alterations are associated with systemic complications including toxicant-induced steatohepatitis [4] and neurotoxicity [42]. Notably, the present study differs from most of the previous research in that it examines the impact of maternal PCB exposure on offspring microbiota. While research is largely limited, Rude and colleagues found that exposure via the maternal diet resulted in a dysbiotic microbial signature [6].Specifically, they observed alterations in offspring beta diversity as a result of maternal PCB exposure. While we did not observe a significant impact of PCB exposure on overall beta diversity measures in the present study, we did find a significant interaction between PCB exposure and exercise in subgroup analysis, and we also observed that PCB exposure effects were masked in the high-fat diet in subgroup analysis (Table 3). However, we did observe that PCB exposure significantly decreased richness and phylogenetic diversity overall (Table 1), and also observed several taxonomic differences by PCB exposure overall. When comparing the present study to Rude et. al, there were no similarities in specific taxa altered as a result of maternal PCB exposure. The differences in our findings compared to Rude et. al. may be due to their use of a different mouse model, our exclusive use of female offspring, use of a different PCB mixture and/or differences in timing of PCB exposure [6]. Nevertheless, this growing body of evidence supports that there is a potential for maternal PCB exposure to impact offspring microbial composition, and thus should be further explored.
In the present study, because some of the 2-way and 3-way interactions for beta diversity measures had a p-value less than 0.10, we conducted exploratory analyses to further examine subgroup-specific effects (Table 3). We observed an interaction between PCB exposure and exercise in mice that were on a high-fat diet for both unweighted and weighted UniFrac distances. We also observed similar interactions between PCB exposure and exercise in mice that were on a regular diet. While exploratory, these findings suggest that there may be important interactions between maternal PCB exposure and exercise on offspring microbial composition that should be examined further in future studies utilizing a larger sample size. In a similar analysis examining the impact of PCB exposure in the four diets + exercise sub-groups, we observed a significant effect of PCB exposure on the microbial composition of mice that were exercised and on a regular diet (weighted and unweighted Unifrac distances). While preliminary, these findings suggest that the effect of maternal PCB exposure may be influenced by the combination of diet and exercise variables, and highlight a need for further exploration.
In the present study we observed several taxa that were significantly different between maternally PCB-exposed or non-exposed offspring. Firstly, Akkermansia muciniphila was significantly reduced as a result of maternal PCB exposure (Figure 3). It should be noted that Verrucomirobiacae and Akkermansia statistically exhibit the same magnitude of response since A. muciniphila is the sole microbe currently classified under that family and genus (Figures 1 and 2). A reduction in A. muciniphila has also been observed by others as a result of direct PCB exposure. For example, Tian and colleagues found that early-life PCB exposure significantly reduced A. muciniphila at 13 weeks after exposure. Interestingly, this group also found that this alteration persisted into adulthood [5]. Of note, A. muciniphila plays a role in the maintenance of the intestinal barrier, has been consistently inversely related to chronic disease risk and inflammation, and may play a role in the maintenance of metabolic health [43-45]. Desulfovibrionaceae spp. were also decreased in the present study as a result of maternal PCB exposure (Figure 3). While little has been observed with this family as a result of persistent organic pollutant exposure, several species within this family have the sulfate-reducing capacity with hydrogen sulfide as a product, which may increase intestinal inflammation and disrupt the intestinal barrier [46, 47]. However, other studies have demonstrated that sulfate-reducing bacteria like Desulfovibrio spp. do not always elicit detrimental impacts and may even have protective effects [48, 49]. In the present study, we also observed that Sutterella sp. was decreased as a result of PCB exposure. This alteration in Sutterella sp. has been observed with pollutant exposure previously. Johanson et al. observed that in a study examining maternal exposure to a mixture of persistent organic pollutants, an unclassified Sutterella sp. was reduced with pollutant exposure [50]. While most microbial groups were found to be reduced with maternal PCB exposure, one group that was higher with PCB exposure was Anaeroplasmataceae, including Anaeroplasma sp. (Figures 1 and 3). Although research is largely lacking, some studies have found an Anaeroplasma sp. to be increased in mice with a diabetic phenotype [51]. More research is needed to determine the main functions of these Anaeroplasma sp. before a clear hypothesis can be made as to why we observed an increase in offspring of maternally PCB-exposed mice.
In the present study, exercise was introduced as a potential modifier of pollutant-induced impact on the microbiome, as our prior research clearly demonstrates the effects of prenatal exercise on matured offspring insulin sensitivity and improves adult offspring glucose homeostasis [28, 52, 53]. Additionally, others have demonstrated that exercise is a probable modulator of the gut microbiome [54, 55] and recent studies have confirmed the link of exercise with the intestinal microbial community [56, 57]. Despite this growing body of research into the developing gut microbiome and dysbiosis, and the well-established benefits of exercise on maternal-child health and perinatal outcomes [58-60], few studies have investigated the impact of maternal exercise before, during and post-pregnancy on the gut microbiome of offspring. Notably, recent studies have found that maternal rats who had greater physical activity during their pregnancy transferred microbes beneficial to gut health to their offspring [19, 27]. In the current study, we did not observe exercise to be protective against pollutant-induced effects on the microbiome, as there were no significant 2-way or 3-way interactions noted for measures of microbial diversity or composition. Yet, from our compositional subgroup analysis we did observe a significant interaction between PCB exposure and exercise irrespective of diet. Future research should explore whether type, during, or timing of exercise intervention impacts any effects that may be exerted on the microbiome.
In addition to maternal exercise, we were also interested if high-fat feeding would exacerbate any observed PCB 126 effects in offspring. High-fat diets have been associated with alterations in alpha diversity, specifically, reductions in richness and evenness [61, 62], and modulate the toxicity of environmental pollutants in multiple mouse models [12, 47-49]. For example, Chi and colleagues showed that mice gut microbiota was significantly impacted by PCB exposure following high-fat diet feeding [63-65]. Additionally, high-fat feeding has been observed to result in an increased F/B ratio [2, 5]. We did not observe any significant interactions between diet and pollutant exposure in the present study, but we observed significant main effects due to diet. The microbial changes observed as a result of diet in the present study were consistent with the known effects of high-fat feeding, including an overall effect on alpha diversity and F/B ratio, with high-fat feeding being associated with lower measures of microbial richness, evenness, and diversity, and a higher F/B ratio [61, 62].
This study had multiple limitations including relatively small sample sizes for the individual groups and the exclusive examination of female offspring. Some studies have demonstrated that female mice appear to be more sensitive to PCB-induced toxicity, but we were unfortunately not able to investigate sex differences herein [66, 67]. Though we detected an overall effect of PCB exposure on gut microbiota, our study was only powered to detect large effect sizes for interactions between PCB and diet or exercise, and these largely failed to be detected. Future studies may consider larger sample sizes to better investigate attenuation by maternal exercise and exacerbation by high-fat diet. Additionally, it is important to note that human exposure to toxicants, like PCB 126, occurs at usually low levels over the life course. Our mouse model of relatively high acute exposures attempts to increase body burdens quickly during the short time of gestation in mice. Unfortunately, blood levels of PCB 126 were not measured in Dams from this current study. Additionally, we acknowledge the limitations of 16S rRNA sequencing for microbial analysis, as it only provides a snapshot of the microbial community and does not allow for a direct understanding of functionality. Similarly, here we could not determine if the microbiota changes are caused by maternal microbiota changes or if the developmental exposures to the PCBs themselves that alter the offspring microbiota. Future studies should focus on utilization of shotgun metagenomics approaches to more accurately investigate species and strain level changes as well as assess functional capacity of the microbial communities. Another limitation of our study is that we cannot determine if effects of maternal PCB exposure in the offspring is due to preconception exposure, in utero exposure, or to exposure during lactation, as there is evidence that dioxin-like PCBs can cross the placental barrier during gestation and excrete in milk during lactation [68, 69]. Future studies utilizing more rigorous cross-fostering strategies will be required to tease out these differences in exposure timing. Nevertheless, the findings of this study contribute to the growing body of evidence demonstrating the detrimental impact of dioxin-like pollutant exposure during critical windows of exposure and development and indicate a need for future research to examine this relationship and the role of lifestyle modifications in the exacerbation or mitigation of PCB-induced effects.
Supplementary Material
Highlights.
- Maternal exposure to PCB 126 decreased microbiome diversity in aged offspring 
- Maternal exposure to PCB 126 impacted depleted offspring Akkermansia muciniphila 
- Maternal exercise or offspring high-fat feeding did not modulate PCB effects 
Funding:
This research was supported in part by the National Institute of Environmental Health Sciences [P30ES020957, R00ES028734, P42ES007380, P30ES026529] and the Office of the Vice President for Research at Wayne State University. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Footnotes
Declaration of interests
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Clemente JC, et al. , The impact of the gut microbiota on human health: an integrative view. Cell, 2012. 148(6): p. 1258–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Petriello MC, et al. , Dioxin-like PCB 126 increases intestinal inflammation and disrupts gut microbiota and metabolic homeostasis. Environ Pollut, 2018. 242(Pt A): p. 1022–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhang L, et al. , Persistent Organic Pollutants Modify Gut Microbiota-Host Metabolic Homeostasis in Mice Through Aryl Hydrocarbon Receptor Activation. Environ Health Perspect, 2015. 123(7): p. 679–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Wahlang B, et al. , Polychlorinated biphenyls altered gut microbiome in CAR and PXR knockout mice exhibiting toxicant-associated steatohepatitis. Toxicol Rep, 2021. 8: p. 536–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tian Y, et al. , Early Life Short-Term Exposure to Polychlorinated Biphenyl 126 in Mice Leads to Metabolic Dysfunction and Microbiota Changes in Adulthood. Int J Mol Sci, 2022. 23(15). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Rude KM, et al. , Developmental exposure to polychlorinated biphenyls (PCBs) in the maternal diet causes host-microbe defects in weanling offspring mice. Environ Pollut, 2019. 253: p. 708–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ribière C, et al. , Oral exposure to environmental pollutant benzo[a]pyrene impacts the intestinal epithelium and induces gut microbial shifts in murine model. Sci Rep, 2016. 6: p. 31027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Uwimana E, et al. , Atropselective Oxidation of 2,2',3,3',4,6'-Hexachlorobiphenyl (PCB 132) to Hydroxylated Metabolites by Human Liver Microsomes and Its Implications for PCB 132 Neurotoxicity. Toxicol Sci, 2019. 171(2): p. 406–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Singleman C, et al. , Toxic Effects of Polychlorinated Biphenyl Congeners and Aroclors on Embryonic Growth and Development. Environ Toxicol Chem, 2021. 40(1): p. 187–201. [DOI] [PubMed] [Google Scholar]
- 10.Simhadri JJ, et al. , Biomarkers of metabolic disorders and neurobehavioral diseases in a PCB- exposed population: What we learned and the implications for future research. Environ Res, 2020. 191: p. 110211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Grimm FA, et al. , Cardiovascular Effects of Polychlorinated Biphenyls and Their Major Metabolites. Environ Health Perspect, 2020. 128(7): p. 77008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Guo W, et al. , Persistent Organic Pollutants in Food: Contamination Sources, Health Effects and Detection Methods. Int J Environ Res Public Health, 2019. 16(22). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Carpenter DO, Polychlorinated biphenyls (PCBs): routes of exposure and effects on human health. Rev Environ Health, 2006. 21(1): p. 1–23. [DOI] [PubMed] [Google Scholar]
- 14.Nash MJ, Frank DN, and Friedman JE, Early Microbes Modify Immune System Development and Metabolic Homeostasis-The “Restaurant” Hypothesis Revisited. Front Endocrinol (Lausanne), 2017. 8: p. 349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kalbermatter C, et al. , Maternal Microbiota, Early Life Colonization and Breast Milk Drive Immune Development in the Newborn. Front Immunol, 2021. 12: p. 683022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Barker DJ, The origins of the developmental origins theory. J Intern Med, 2007. 261(5): p. 412–7. [DOI] [PubMed] [Google Scholar]
- 17.Gluckman PD, et al. , Effect of in utero and early-life conditions on adult health and disease. N Engl J Med, 2008. 359(1): p. 61–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Choi JJ, et al. , Exercise attenuates PCB-induced changes in the mouse gut microbiome. Environ Health Perspect, 2013. 121(6): p. 725–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zhou L, et al. , Maternal Exercise Improves High-Fat Diet-Induced Metabolic Abnormalities and Gut Microbiota Profiles in Mouse Dams and Offspring. Front Cell Infect Microbiol, 2020. 10: p. 292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lefever DE, et al. , TCDD modulation of gut microbiome correlated with liver and immune toxicity in streptozotocin (STZ)-induced hyperglycemic mice. Toxicol Appl Pharmacol, 2016. 304: p. 48–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ley RE, et al. , Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A, 2005. 102(31): p. 11070–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li QQ, et al. , Persistent organic pollutants and adverse health effects in humans. J Toxicol Environ Health A, 2006. 69(21): p. 1987–2005. [DOI] [PubMed] [Google Scholar]
- 23.Maher SE, et al. , The association between the maternal diet and the maternal and infant gut microbiome: a systematic review. Br J Nutr, 2020: p. 1–29. [DOI] [PubMed] [Google Scholar]
- 24.Mirpuri J, Evidence for maternal diet-mediated effects on the offspring microbiome and immunity: implications for public health initiatives. Pediatr Res, 2021. 89(2): p. 301–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lundgren SN, et al. , Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome, 2018. 6(1): p. 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hasebe K, et al. , The influence of maternal unhealthy diet on maturation of offspring gut microbiota in rat. Anim Microbiome, 2022. 4(1): p. 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bhagavata Srinivasan SP, et al. , Impacts of Diet and Exercise on Maternal Gut Microbiota Are Transferred to Offspring. Front Endocrinol (Lausanne), 2018. 9: p. 716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Carter LG, et al. , Perinatal exercise improves glucose homeostasis in adult offspring. Am J Physiol Endocrinol Metab, 2012. 303(8): p. E1061–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Rice BB, et al. , Lack of Offspring Nrf2 Does Not Exacerbate the Detrimental Metabolic Outcomes Caused by In Utero PCB126 Exposure. Front Endocrinol (Lausanne), 2021. 12: p. 777831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rashid CS, et al. , Perinatal Polychlorinated Biphenyl 126 Exposure Alters Offspring Body Composition. J Pediatr Biochem, 2013. 3(1): p. 47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Fujimura KE, et al. , Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat Med, 2016. 22(10): p. 1187–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cohen J, A power primer. Psychol Bull, 1992. 112(1): p. 155–9. [DOI] [PubMed] [Google Scholar]
- 33.Kembel SW, et al. , Picante: R tools for integrating phylogenies and ecology. Bioinformatics, 2010. 26(11): p. 1463–4. [DOI] [PubMed] [Google Scholar]
- 34.Oksanen J, S.G., Blanchet F, Kindt R, Legendre P, Minchin P,, et al. _vegan:Community Ecology Package_. R package version 2.6-4,. 2022; Available from: <https://CRAN.R-project.org/package=vegan>. [Google Scholar]
- 35.McMurdie PJ and Holmes S, phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One, 2013. 8(4): p. e61217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lozupone CA, et al. , Quantitative and qualitative beta diversity measures lead to different insights into factors that structure microbial communities. Appl Environ Microbiol, 2007. 73(5): p. 1576–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Love MI, Huber W, and Anders S, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol, 2014. 15(12): p. 550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Dong TS and Gupta A, Influence of Early Life, Diet, and the Environment on the Microbiome. Clin Gastroenterol Hepatol, 2019. 17(2): p. 231–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ahn J and Hayes RB, Environmental Influences on the Human Microbiome and Implications for Noncommunicable Disease. Annu Rev Public Health, 2021. 42: p. 277–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mohr AE, et al. , The athletic gut microbiota. J Int Soc Sports Nutr, 2020. 17(1): p. 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jin Y, et al. , Effects of environmental pollutants on gut microbiota. Environ Pollut, 2017. 222: p. 1–9. [DOI] [PubMed] [Google Scholar]
- 42.Rude KM, Keogh CE, and Gareau MG, The role of the gut microbiome in mediating neurotoxic outcomes to PCB exposure. Neurotoxicology, 2019. 75: p. 30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yoon HS, et al. , Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat Microbiol, 2021. 6(5): p. 563–573. [DOI] [PubMed] [Google Scholar]
- 44.van der Lugt B, et al. , Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1 (−/Δ7) mice. Immun Ageing, 2019. 16: p. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Rodrigues VF, et al. , Akkermansia muciniphila and Gut Immune System: A Good Friendship That Attenuates Inflammatory Bowel Disease, Obesity, and Diabetes. Front Immunol, 2022. 13: p. 934695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kushkevych I, Dordević D, and Kollár P, Analysis of Physiological Parameters of Desulfovibrio Strains from Individuals with Colitis. Open Life Sci, 2018. 13: p. 481–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Figliuolo VR, et al. , Sulfate-reducing bacteria stimulate gut immune responses and contribute to inflammation in experimental colitis. Life Sci, 2017. 189: p. 29–38. [DOI] [PubMed] [Google Scholar]
- 48.Hong Y, et al. , Desulfovibrio vulgaris, a potent acetic acid-producing bacterium, attenuates nonalcoholic fatty liver disease in mice. Gut Microbes, 2021. 13(1): p. 1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen YR, et al. , Desulfovibrio is not always associated with adverse health effects in the Guangdong Gut Microbiome Project. PeerJ, 2021. 9: p. e12033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Johanson SM, et al. , Maternal exposure to a human relevant mixture of persistent organic pollutants reduces colorectal carcinogenesis in A/J Min/+ mice. Chemosphere, 2020. 252: p. 126484. [DOI] [PubMed] [Google Scholar]
- 51.Brown K, et al. , Prolonged antibiotic treatment induces a diabetogenic intestinal microbiome that accelerates diabetes in NOD mice. Isme j, 2016. 10(2): p. 321–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Platt KM, et al. , Effects of maternal controlled exercise on offspring adiposity and glucose tolerance. J Dev Orig Health Dis, 2022. 13(4): p. 455–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Carter LG, et al. , Maternal exercise improves insulin sensitivity in mature rat offspring. Med Sci Sports Exerc, 2013. 45(5): p. 832–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Mailing LJ, et al. , Exercise and the Gut Microbiome: A Review of the Evidence, Potential Mechanisms, and Implications for Human Health. Exerc Sport Sci Rev, 2019. 47(2): p. 75–85. [DOI] [PubMed] [Google Scholar]
- 55.Barton W, et al. , The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut, 2018. 67(4): p. 625–633. [DOI] [PubMed] [Google Scholar]
- 56.Motiani KK, et al. , Exercise Training Modulates Gut Microbiota Profile and Improves Endotoxemia. Med Sci Sports Exerc, 2020. 52(1): p. 94–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Liu Y, et al. , Gut Microbiome Fermentation Determines the Efficacy of Exercise for Diabetes Prevention. Cell Metab, 2020. 31(1): p. 77–91.e5. [DOI] [PubMed] [Google Scholar]
- 58.Prather H, Spitznagle T, and Hunt D, Benefits of exercise during pregnancy. Pm r, 2012. 4(11): p. 845–50; quiz 850. [DOI] [PubMed] [Google Scholar]
- 59.Piercy KL, et al. , The Physical Activity Guidelines for Americans. Jama, 2018. 320(19): p. 2020–2028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.da Silva SG, et al. , Leisure-Time Physical Activity in Pregnancy and Maternal-Child Health: A Systematic Review and Meta-Analysis of Randomized Controlled Trials and Cohort Studies. Sports Med, 2017. 47(2): p. 295–317. [DOI] [PubMed] [Google Scholar]
- 61.Wan Y, et al. , Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: a 6-month randomised controlled-feeding trial. Gut, 2019. 68(8): p. 1417–1429. [DOI] [PubMed] [Google Scholar]
- 62.Murphy EA, Velazquez KT, and Herbert KM, Influence of high-fat diet on gut microbiota: a driving force for chronic disease risk. Curr Opin Clin Nutr Metab Care, 2015. 18(5): p. 515–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Klinge CM, et al. , Combined exposure to polychlorinated biphenyls and high-fat diet modifies the global epitranscriptomic landscape in mouse liver. Environ Epigenet, 2021. 7(1): p. dvab008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hennig B, et al. , Dietary fat interacts with PCBs to induce changes in lipid metabolism in mice deficient in low-density lipoprotein receptor. Environ Health Perspect, 2005. 113(1): p. 83–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Chi Y, et al. , PCBs-high-fat diet interactions as mediators of gut microbiota dysbiosis and abdominal fat accumulation in female mice. Environ Pollut, 2018. 239: p. 332–341. [DOI] [PubMed] [Google Scholar]
- 66.Wahlang B, et al. , Identifying sex differences arising from polychlorinated biphenyl exposures in toxicant-associated liver disease. Food Chem Toxicol, 2019. 129: p. 64–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gang N, et al. , Sex-specific Associations Between Type 2 Diabetes Incidence and Exposure to Dioxin and Dioxin-like Pollutants: A Meta-analysis. Front Toxicol, 2021. 3: p. 685840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zetterström R, Child health and environmental pollution in the Aral Sea region in Kazakhstan. Acta Paediatr Suppl, 1999. 88(429): p. 49–54. [DOI] [PubMed] [Google Scholar]
- 69.Lundqvist C, et al. , The effects of PCBs and dioxins on child health. Acta Paediatr Suppl, 2006. 95(453): p. 55–64. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



