Endocrine disruptor-induced epimutagenesis in vitro: Insight into molecular mechanisms

  1. Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio

Editors

  • Reviewing Editor
    Isabelle Mansuy
    University/ETH Zurich, Zurich, Switzerland
  • Senior Editor
    Wei Yan
    The Lundquist Institute, Torrance, United States of America

Reviewer #1 (Public Review):

Summary:

The goal of this study was to use in vitro cell populations to determine mechanisms that may be important for the propagation of epimutations induced by EDCs in vivo. To do this, authors exposed induced pluripotent stem cells (iPS), somatic cells (Sertoli, granulosa), and primordial germ cell like cells (PGCLCs) to BPS, and conducted epigenomic and transcriptomic analyses on outcomes. The importance of estrogen receptors, and the relationship of epigenomic results to genomic sites expressing EREs, were also determined in the different cell types. Results revealed differential effects of BPS in each cell population on each of these endpoints, and that epimutations were prevalent in enhancer regions with EREs with the exception of PGCLCs (which do not express ERs). The authors speculate that because epimutations also occurred in regions without EREs, especially in PGCLCs, other mechanisms may be in place. Finally, epimutations induced in iPSCs exposed to BPS that were subsequently differentiated into PGCLCs demonstrated that most epimutations were corrected.

Strengths:

A strength of this work is the use of different cell types representing somatic cells that would be the major recipient of EDC exposure; pluripotent cells representing preimplantation embryos; and PGCLCs that model the early germline in which epigenetic reprogramming takes place. Work differentiating the iPSCs from PGCLCs with or without BPS exposure at the iPSC level is also very informative as it suggests that most epimutations are corrected, at least in vitro. The paper is well-written and studies were technically well-executed and validated. Results are novel and likely to be of interest to those interested in transgenerational inheritance of environmentally-induced traits, as well as others more broadly interested in epigenetic mechanisms.

Critique/Weaknesses:

(1) A problem with in vitro work is that homogeneous cell lines/cultures are, by nature, absent from the rest of the microenvironment. The authors need to discuss this.

(2) What are n's/replicates for each study? Were the same or different samples used to generate the data for RNA sequencing, methylation beadchip analysis, and EM-seq? This clarification is important because if the same cultures were used, this would allow comparisons and correlations within samples.

(3) In Figure 1, it is interesting that the 50 uM BPS dose mainly resulted in hypermethylation whereas 100 uM appears to be mainly hypomethylation. (This is based on the subjective appearance of graphs). The authors should discuss and/or present these data more quantitatively. For example, what percentage of changes were hypo/hypermethylation for each treatment? How many DMRs did each dose induce? For the RNA-seq results, again, what were the number of up/down-regulated genes for each dose?

(4) Also in Figure 1, were there DMRs or genes in common across the doses? How did DMRs relate to gene expression results? This would be informative in verifying or refuting expectations that greater methylation is often associated with decreased gene expression.

(5) In Figure 2, was there an overlap in the hypo- and/or hyper-methylated DMCs? Please also add more description of the data in 2b to the legend including what the dot sizes/colors mean, etc. Some readers (including me) may not be familiar with this type of data presentation. Some of this comes up in Figure 4, so perhaps allude to this earlier on, or show these data earlier.

(6) iPSCs were derived from male mice MEFs, and subsequently used to differentiate into PGCLCs. The only cell type from an XX female is the granulosa cells. This might be important, and should be mentioned and its potential significance discussed (briefly).

(7) EREs are only one type of hormone response element. The authors make the point that other mechanisms of BPS action are independent of canonical endocrine signaling. Would authors please briefly speculate on the possibility that other endocrine pathways including those utilizing AREs or other HREs may play a role? In other words, it may not be endocrine signaling independent. The statement that the differences between PGCLCs and other cells are largely due to the absence of ERs is overly simplistic.

(8) Interpretation of data from the GO analysis is similarly overly simplistic. The pathways identified and discussed (e.g. PI3K/AKT and ubiquitin-like protease pathways are involved in numerous functions, both endocrine and non-endocrine. Also, are the data shown in Figure 6a from all 4 cell types? I am confused by the heatmap in 6c, which genes were significantly affected by treatment in which cell types?

(9) In Figure 7, what were the 138 genes? Any commonalities among them?

(10) The Introduction is very long. The last paragraph, beginning line 105, is a long summary of results and interpretations that better fit in a Discussion section.

(11) Provide some details on husbandry: e.g. were they bred on-site? What food was given, and how was water treated? These questions are to get at efforts to minimize exposure to other chemicals.

Reviewer #2 (Public Review):

Summary:

This manuscript uses cell lines representative of germ line cells, somatic cells, and pluripotent cells to address the question of how the endocrine-disrupting compound BPS affects these various cells with respect to gene expression and DNA methylation. They find a relationship between the presence of estrogen receptor gene expression and the number of DNA methylation and gene expression changes. Notably, PGCLCs do not express estrogen receptors and although they do have fewer changes, changes are nevertheless detected, suggesting a nonconical pathway for BPS-induced perturbations. Additionally, there was a significant increase in the occurrence of BPS-induced epimutations near EREs in somatic and pluripotent cell types compared to germ cells. Epimutations in the somatic and pluripotent cell types were predominantly in enhancer regions whereas that in the germ cell type was predominantly in gene promoters.

Strengths:

The strengths of the paper include the use of various cell types to address the sensitivity of the lineages to BPS as well as the observed relationship between the presence of estrogen receptors and changes in gene expression and DNA methylation.

Weaknesses:

The weaknesses include the lack of reporting of replicates, superficial bioinformatic analysis, and the fact that exposures are more complicated in a whole organism than in an isolated cell line.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation