Introduction

Obesity and diabetes are chronic metabolic diseases affecting human populations worldwide. These metabolic disorders have increased to unprecedented levels over the past several decades. In the United States, nearly 40% of adults are obese and 7.7% are morbidly obese1. Also, children and adolescents ages 2–19 are affected, with 18.5% obese and 5.6% morbidly obese1. Body mass index (BMI) rose 2.3 kg/m2 from 1988 to 20062,3. Rates of diabetes and metabolic syndrome have also increased and are often coupled with obesity4,5. Caloric imbalance (greater intake/lower expenditure) and genetics are important drivers of obesity. Despite increased efforts in nutrition and fitness education, obesity rates fail to decline and are estimated to reach 51% of the US population by 20306. However, increasing numbers of obesity cases cannot be attributed purely to genetics. GIANT consortium analyses indicate that only 2.7% of increases in BMI can be ascribed to genetics and that the total genetic contribution will only reach 20% once all such genes are identified7,8. Such “missing heritability” cases have been attributed to gene-by-environment interactions9,10. For example, familial heritability and obesity-discordant twins studies and complex trait analyses11,12 indicate that combinations of polymorphisms can account for at most 30–37% of the variance associated with BMI13,14. Environmental exposures, especially during development, have gained recognition as playing important roles in obesity and metabolic syndrome15. Developmental exposures that impact obesity include stress, diet, pollutants, pharmaceuticals and personal care products16,17,18,19. Chemicals that promote obesity and metabolic syndrome have been termed ‘obesogens’20,21. Obesogens can drive obesity by multiple molecular mechanisms. The most researched mechanism involves obesogens acting as agonists of the nuclear receptor PPARγ. Furthermore, hormone receptor assays are often utilized for the in vitro identification of potential obesogens, prior to in vivo validation10. Published work from our group identified the anionic surfactant, dioctyl sodium sulfosuccinate (DOSS), as a probable obesogen using in silico modeling, in vitro receptor ligand binding and transactivation assays and murine preadipocyte adipogenic differentiation22. DOSS (CAS #577-11-7) is also known as AOT, Aerosol OT, diethylhexyl sodium sulfosuccinate, Colace, Docusate sodium, and many other names. Structurally similar obesogens have been classified in literature as PPARγ agonists, including the anionic surfactants sodium dodecyl sulfate (SDS) and sodium dodecylbenzenesulfonate (SDBS), which suggest that surfactants/dispersants constitute a previously unidentified and unexplored class of obesogens23. Epidemiological studies previously identified positive correlations between chemicals measured during pregnancy (e.g. phthalates and persistent organic pollutants) and childhood obesity24,25,26. Exposures during development can profoundly impact epigenetic patterning that persistently impact later life health trajectories27,28,29. As with the other surfactant PFOA, DOSS will be evaluated for its obesogenic potential in vivo during development30. Pregnancy associated exposure may result from other possible sources of DOSS, including dietary emulsifying agents (e.g. colored/flavored powdered beverages) and personal care products31,32,33,34. Approximately 11–38% of pregnant women experience constipation during pregnancy35, and DOSS in the form of Colace/Docusate stool softener is the treatment of choice, being prescribed up to 500 mg/day36,37. In addition, nearly 2 million gallons of COREXIT dispersants, containing DOSS as a principle component, were used to clean up the Deepwater Horizon oil spill.

In the current study, C57BL/6 mouse dams were exposed to DOSS following a dose and exposure scenario relevant to Colace/Docusate stool softener use by pregnant women. The C57BL/6 mouse line was chosen as it is commonly used for diet-induced obesity studies. The offspring of exposed dams were examined using markers of adiposity and metabolic syndrome and a combination of morphometrics (e.g. body composition, circulating adipokines and cytokines, tissue gene expression, glucose tolerance and epigenetic marks such as DNA methylation). This study followed the precedent set by other in vivo studies, which evaluated obesogens that act as ligands to activate PPARγ, including the phthalate metabolite MEHPFull size image

The statistically significant reduction in normalized adiponectin levels observed in treated animals could be due to significant changes in white adipose tissue depots expressing AdipoQ, the gene encoding adiponectin (Supp. Table 1). Consistent with this, a statistically significant down regulation of AdipoQ expression (p = 0.015) was observed in F1 male offspring IWAT tissues from treated versus control dams (Fig. 3C). Although not statistically significant, decreased gene expression of AdipoQ was observed in male gonadal (EWAT) adipose tissue as well (data not shown). These data further support the hypothesis that DOSS exposure during pregnancy results in decreased adiponectin in adult male offspring at 16 weeks. A non-statistically significant trend for lower AdipoQ expression was observed in adult female offspring IWAT of treated dams (data not shown). These results suggest that no change in circulating adipokine levels in females would be observed, further supporting the association between DOSS exposure, adipokine levels and overweight phenotype in male, but not female animals.

Leptin, an adipokine that regulates satiety and appetite, has been shown to be positively correlated with adiposity and obesity49. While other fat depots are capable of producing and secreting more leptin, models based on circulating leptin levels are likely more reliable than IWAT gene expression for whole body response to satiety and appetite. Therefore, this study provided circulating leptin levels. Significantly higher levels of circulating leptin (p = 0.033) were observed in adult F1 males from DOSS-treated dams compared to controls (Fig. 4A). A positive relationship between leptin and adiposity markers is well documented in both humans and mice50,51. Our work demonstrates this same trend as circulating leptin levels were positively correlated with fat mass for DOSS treatment (Fig. 4B). Although some treatment F1 males displayed higher expression of the leptin gene in IWAT relative to controls, the differences were not statistically significant (Fig. 4C).

Figure 4
figure 4

Developmental DOSS treatment alters circulating levels of leptin in male F1 mice. At time of sacrifice (16 weeks), plasma and adipose tissue was collected (n = 12/group). Circulating levels of leptin were determined using MSD assays. Gene expression was determined via RNA isolation, cDNA conversion and qPCR using the delta delta Ct method with Hprt as the housekee** gene. Results are shown for (A) normalized circulating levels of leptin, (B) leptin correlated with fat mass, and (C) leptin gene expression in IWAT (n = 12/group). Graph bars represent means and standard deviations. Linear regression was used to determine correlations between adiponectin and leptin and fat mass (*p < 0.05 unpaired t-test).

DOSS treatment in dams produces a persistent inflammatory state in adult F1 male offspring

Obesity and type 2 diabetes (T2D) are often referred to as a chronic inflammatory diseases of adipose tissues, which are presumably driven by infiltration of adipose tissue with macrophages52. IL-6 is a proinflammatory cytokine secreted by immune cells. Circulating IL-6 levels were measured to determine if offspring of DOSS-treated animals showed indications of such an inflammatory state. Significantly higher circulating levels of IL-6 in treatment (48.12 pg/mL) versus control (13.70 pg/mL) adult F1 males were observed at p = 0.011 (Fig. 5A). Unlike adiponectin and leptin, a correlation between adipose tissue weight and circulating IL-6 levels was not observed in either treatment or control animals (Supp. Fig. 2A). IL-6 gene expression changes were also monitored in IWAT adipose tissue for potential associations with inflammation. No statistically significant differences in IL-6 gene expression were observed between treatment and control animals (Supp. Fig. 2B) despite increased circulating levels of IL-6 in treatment animals (Fig. 5A). This finding may suggest the contribution of other immune cell/fat depots to overall circulating IL-6 levels. Within adipose tissue, the stromal vascular fraction accounts for 90% of the IL-6 expression in adipose tissue even though it represents a small percentage of adipose tissue cells53.

Figure 5
figure 5

Developmental DOSS treatment promotes a proinflammatory state in adult F1 male mice. At time of sacrifice (16 weeks), plasma and adipose tissue were collected (n = 12/group). Circulating levels of IL-6 and TNF-α were determined using MSD assays, gene expression using RNA isolation, along with cDNA conversion and qPCR using the delta delta Ct method with Hprt as the housekee** gene. Results are shown for (A) circulating levels of IL-6 in plasma, (B) circulating levels of TNF-α in plasma, (C) Cox2 gene expression in IWAT tissue, and (D) Nox4 gene expression in IWAT tissue (*p < 0.05 unpaired t-test). Some data points were excluded via outlier testing. Graph bars represent means and standard deviations.

TNFα is another inflammatory cytokine secreted by immune cells and has been shown to be positively correlated with obesity and T2D. Therefore, mouse plasma were analyzed for levels of TNFα. The mean circulating TNFα was higher in treatment adult F1 males (8.44 pg/mL) compared to controls (7.54 pg/mL), although not statistically significant (Fig. 5B).

Other genes investigated included cyclooxygenase 2 (Cox2) and NADPH oxidase (Nox4; Supp. Table 1). Elevated expression of Cox2 and Nox4 are associated with the presence of reactive oxygen species and systemic chronic inflammation54,55. Both Cox2 and Nox4 expressions were significantly increased in IWAT for DOSS-treatment versus control adult F1 males (Fig. 5C,D). Thus, the increased expression of multiple inflammatory markers in this work suggests that a state of chronic inflammation exists in offspring of DOSS-treated dams.

DOSS treatment in dams produce altered DNA promoter methylation of IL-6 and Cox2 genes in adult F1 male offspring

Increased levels of gene expression (Cox2 and Nox4) and circulating levels of IL-6, adipokines and cytokines were observed in 16 week-old F1 males of DOSS treated dams. These findings suggest alterations of promoter methylation at these loci in IWAT. Therefore, promoter methylation was investigated at these loci, as well as at previously identified gene promoters reported in the literature to be regulated by DNA methylation in response to high-fat diet, obesity or T2D (Leptin, Pparg, Glut4, Fasn, Irs1, Hmox1, Fabp4). A complete list of loci, bisulfite primers and references used in this work are presented in Supp. Table 2.

The promoter region of the IL-6 gene in IWAT tissue was investigated for changes in DNA methylation. Five previously identified CpG sites roughly 300 bp upstream of the IL-6 transcription start site were evaluated for methylation status56. Results in Supp. Fig. 3A show DOSS treatment to be associated with significantly reduced methylation specifically at CpG site 2 by two-way ANOVA (p = 0.004) and by Sidak’s post hoc test (p = 0.045). Although there was no statistically significant correlation (p = 0.705) between IWAT gene expression and DNA methylation at CpG site 2 (Supp. Fig. 4A), there was a statistically significant inverse correlation (p = 0.011) between IL-6 CpG site 2 methylation and circulating IL-6 levels. (Supp. Fig. 3B). To the best of our knowledge this is the first significant finding of IL-6 CpG site 2 methylation status in adipose tissue as it relates to circulating IL-6 levels, though it’s biological importance remains to be determined. Although multiple tissue sources of IL-6 contribute to circulating levels, the potential of CpG site 2 to serve as a biomarker for the persistent effects in offspring of DOSS exposed mothers warrants further investigation.

Changes in Cox2 promoter hypermethylation upon exposure have been linked to Cox2 gene expression in other studies57. Therefore, fourteen CpG sites approximately 500 bp upstream of the Cox2 transcription start site were evaluated for methylation status. Results demonstrate that although DOSS treatment was not significantly associated (p = 0.128) with Cox2 promoter methylation in this region (Supp. Fig. 3C), average DNA methylation at CpG site 1 was significantly reduced (p = 0.005) in treatment males (Supp. Fig. 3D). However, there was only a modest correlation (p = 0.280) between CpG site 1 methylation and Cox2 gene expression (Supp. Fig. 4B).

Bisulfite sequencing for six CpG sites in the adiponectin promoter were targeted (i.e. four adiponectin region 2 sites, associated with high-fat diet induced gene expression, and two nearby adiponectin region 1 sites having no previously documented associations with gene expression). Statistically significant hypomethylation was observed in region 1, particularly at CpG site 1 (Supp. Fig. 4C) in tissues from treatment males. There was no statistically significant difference in region 2 promoter methylation between treatment and controls. Decreased levels of circulating adiponectin and gene expression in IWAT tissue suggest the possible hypermethylation of the adiponectin promoter at each CpG site in region 2, resulting in repression of gene expression upon DOSS treatment (Supp. Fig. 4D). Region 2 CpG site 3 showed that 58% of DOSS treatment animals had over 80% methylation compared to 29% of controls with over 80% methylation (Supp. Fig. 4E). However, no statistically significant correlations were observed between region 2 CpG site 3 methylation and gene expression (Supp. Fig. 4E). Furthermore, no statistically significant differences in promoter methylation were observed for Leptin, Fasn, Pparg, Glut4, Irs1, Fabp4, or Hmox1 (Supp. Table 3).

DOSS treatment in dams induces glucose intolerance in adult F1 male offspring

Glucose tolerance tests are used clinically to assess metabolic function in humans at risk for diabetes and experimentally in obesity and diabetic mouse models. To determine the effect of maternal DOSS treatment during development on glucose tolerance in adult offspring, oral glucose tolerance tests were performed at 12 weeks of age on offspring of control and DOSS-treated dams. Results demonstrate that only male offspring of DOSS treated dams exhibit marked glucose intolerance (Fig. 6). Specifically, Fig. 6A shows statistically significant higher blood glucose levels at 30 minutes in male offspring of DOSS treated vs. control dams. Similarly, these DOSS treatment F1 males show a significantly higher overall glucose tolerance impairment (p = 0.010), as indicated by the area under the curve (AUC) for the entire time course (Fig. 6B). No specific time points or AUC differences were observed amongst the female offspring (Fig. 6C,D). Male-specific results are commonly observed in C57BL/6 mouse models for high-fat diet induced obesity58,59.

Figure 6
figure 6

Developmental DOSS treatment in dams promotes glucose intolerance, and alters expression and circulating levels of insulin in adult F1 male offspring. Pregnant C57BL/6J dams were treated with either vehicle control or DOSS from E11.5 through weaning. F1 pups were assessed for indications of metabolic syndrome at 12 weeks using oral glucose tolerance testing. Basal glucose measurements were taken after fasting for 6 hours. Mice were then administered a 2 mg/g bolus of glucose and blood glucose was measured at 15, 30, 60 and 120 minutes. Blood glucose measurements for all male (n = 22/group) mice are shown in (A) with corresponding area under the curve values in (B). Blood glucose measurements for all female (n = 16/group) mice are shown in (C). Blood glucose area under the curve values for females are shown in (D). Two-Way Anova was used to determine significant differences in blood glucose at different time points (*p < 0.05). Area under the curve values were calculated for each mouse, pooled and student’s t-tests were used to determine significance (**p < 0.01). Plasma and adipose tissue were collected at time of sacrifice. Circulating levels of C-peptide were determined using Alpco ELISA assays. Linear regression was used to determine correlations between adiponectin, adipose weight and glucose tolerance. Results are shown for (E) fasting and glucose stimulated C-peptide levels at 12weeks, (F) raw adiponectin correlated with fat mass, (G) adipose tissue weight correlated with oGTT AUC as a measure of glucose tolerance, (H) normalized adiponectin correlated with oGTT AUC as a measure of glucose tolerance (*p < 0.05 unpaired t-test, control vs. DOSS treatment; #p < 0.05 unpaired t-test, fasting vs. glucose stimulated C peptide). Graph bars represent means and standard deviations.

C-reactive peptide (C-peptide), a more stable marker for insulin due to its longer half-life, is released in a 1:1 ratio to insulin, making it more conducive for plasma studies with variable storage times and conditions. Therefore, circulating C-peptide levels were measured as a stable surrogate for insulin secretion in male offspring (n = 10/group) to determine whether glucose intolerance in DOSS treatment F1 males resulted from insulin desensitization marked by hyperinsulinemia. Significantly higher levels of fasting C-peptide (p = 0.040) was measured in male offspring of DOSS-treated dams (718 pM) relative to controls (563 pM; Fig. 6E). Furthermore, upon glucose stimulation, a statistically significant increase (p = 0.020) in C-peptide levels was observed only in control animals (Fig. 6E). These results suggest that adult male offspring of DOSS-treated dams demonstrate hyperinsulinemia and blunted insulin responsiveness to glucose stimulation. Primarily glucose intolerance and adiponectin, but also obesity and adiponectin have been described to have an inverse relationship60, while positive correlations between circulating adiponectin and adipose fat mass have been observed61,62. Consistent with these results, raw adiponectin level and glucose intolerance are positively correlated with adipose tissue mass in our animals (Fig. 6F,G), and adiponectin levels are negatively correlated with glucose intolerance (Fig. 6H).

DOSS treatment in dams produces adult F1 male offspring with elevated circulating phospholipid patterns commonly observed in long-term high-fat diet induced obesity and diabetes

Due to recent advancements in mass spectrometry (MS) and ultra-high performance liquid chromatography (UHPLC) untargeted lipidomic applications have broadened to investigate lipid profiles associated with obesity and diabetes63,64,65. Biomarkers identified using this approach have the potential to elucidate cellular mechanisms associated with disease progression. Therefore, an untargeted lipidomics workflow was incorporated to identify significant and persistent changes in plasma lipid profiles as a result of DOSS exposure during development. Overall, 664 features were identified in positive MS scan mode and 171 features identified in negative MS scan mode. Various phospholipids (e.g. phosphatidyl choline (PC) and phosphatidylethanolamine (PE)) and sterol lipid (e.g. cholesterol ester (CE)) species were increased in 16 week-old F1 male offspring of DOSS-treated dams. As shown in Fig. 7A–H, the following lipid species demonstrated statistically significant or near significant increases in relative peak areas: PC(34:3) (p = 0.043), PC(36:1) (p = 0.005), PC(38:4) (p = 0.012), PC(18:0_20:1) (p = 0.018), PC(18:0_20:3) (p = 0.038), PE(18:0_20:1) (p = 0.015), PE(18:0_20:3) (p = 0.044), and CE(20:3) (p = 0.058). Several of the lipid species annotated in this work have also been highlighted in literature66 as putative biomarkers for high-fat diet induced obesity in C57BL/6 mice, including PC(36:1), PC(38:3), PC(38:4) and CE(20:3). Lysophosphatidylcholine (LPC) lipid species have been shown to be reduced in response to high-fat diet, obesity and diabetes67. Reductions in several novel LPCs were observed in response to DOSS exposure during development, including LPC 15:0, 17:0, 19:0, 20:0, 22:0, 22:1 and 24:1. These results suggest that developmental exposure to DOSS followed by a normal ad lib diet promotes persistent dyslipidemia akin to long-term high fat-diet induced obesity in adult mice.

Figure 7
figure 7

DOSS treatment in dams promotes persistent changes in circulating phospholipids in adult F1 male offspring. At time of sacrifice plasma was collected from F1 animals (n = 12/group). Untargeted lipidomics was used to determine the presence of circulating lipids. Lipid Match was used to identify lipid species based on unique m/z ratios and retention times. Relative peak area was calculated in FRAMe. Relative peak areas between DOSS treated and vehicle control F1 males are shown for (A) phosphatidyl choline PC(34:3), (B) PC(36:1), (C) PC(38:4), (D) PC(18:0_20:1), (E) PC(18:0_20:3), (F) phosphatidylethanolamine PE(18:0_20:1), (G) PE(18:0_20:3), and (H) cholesterol ester CE(20:3) (*p < 0.05 **p < 0.01 unpaired t-test control vs. DOSS treatment). Some data points were excluded via outlier testing. Graph bars represent means and standard deviations.