Abstract

Persistent exposure to man-made endocrine disrupting chemicals during fetal endocrine development may lead to disruption of metabolic homeostasis contributing to childhood obesity. Limited cellular platforms exist to test endocrine disrupting chemical-induced developmental abnormalities in human endocrine tissues. Here we use an human-induced pluripotent stem cell-based platform to demonstrate adverse impacts of obesogenic endocrine disrupting chemicals in the developing endocrine system. We delineate the effects upon physiological low-dose exposure to ubiquitous endocrine disrupting chemicals including, perfluoro-octanoic acid, tributyltin, and butylhydroxytoluene, in endocrine-active human-induced pluripotent stem cell-derived foregut epithelial cells and hypothalamic neurons. Endocrine disrupting chemicals induce endoplasmic reticulum stress, perturb NF-κB, and p53 signaling, and diminish mitochondrial respiratory gene expression, spare respiratory capacity, and ATP levels. As a result, normal production and secretion of appetite control hormones, PYY, α-MSH, and CART, are hampered. Blocking NF-κB rescues endocrine disrupting chemical-induced aberrant mitochondrial phenotypes and endocrine dysregulation, but not ER-stress and p53-phosphorylation changes.

Highlights

  • D endocrine disrupting chemicals in the developing endocrine system

  • The transcriptional we explored whether blocking NF-κB pathway with the cell regulation of mitochondrial respiratory genes, SCO2, POLRMT, permeable NF-κB peptide inhibitor (NF-κBi), SN50, can rescue TFAM, and CYTB5 were partially restored upon NF-κBi these phenotypes

  • Upon co-treatment with EDCs and it is important to note that NF-κBi treatment did not NF-κBi in the iForegut epithelium (iFGE) and iPSC. The neuropeptidergic hypothalamic neurons (iHTNs), we found an overall decrease in rescue changes in endoplasmic reticulum (ER) stress proteins, IRE1, Ero[1], and BiP (Supphospho-p65, canonical (p50/p105), and non-canonical (p52/ plementary Fig. 10a), suggesting that ER stress may be upstream p100) pathway almost returning to the levels of the vehicle of the NF-κB pathway perturbations (Fig. 7)

Read more

Summary

Results

Non-integrating reprogramming of peripheral blood mononuclear cells (PBMCs) to iPSCs was performed using the episomal (OriP/EBNA1) plasmid-based method similar to published protocols in our lab[36, 37] This included nuclear transfection of seven transient and episomally expressed reprogramming factors OCT3/4, SOX2, KLF4, LIN28, non-transforming L-MYC, SV40 large T antigen (SV40LT), and shRNA against TP53 (Supplementary Fig. 1a). Representative images from independent iPSC lines used in this study (80iCTR Tn2 and 201iCTR NTn4) exhibited typical features of hPSCs such as tight colonies with high nucleus to cytoplasm ratio (Supplementary Fig. 1b) In this manuscript, we show ability to culture gut organoids as They exhibit robust alkaline phosphatase activity, and adherent monolayers of gut epithelium, making them strong expression of nuclear (OCT3/4, NANOG, SOX2) and amenable to toxicology and endocrine function assays.

D SYP E f
40 GABA j
D Vh PFOA TBT BHT Comb IRE1
NF-κB p65
Basal respiration
C NF-κB perturbation p53 perturbation
Discussion
E Author contributions
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.