Deficiency of the oxidative stress-responsive kinase p70S6K1 restores autophagy and ameliorates neural tube defects in diabetic embryopathy

Am J Obstet Gynecol. 2020 Nov;223(5):753.e1-753.e14. doi: 10.1016/j.ajog.2020.05.015. Epub 2020 May 13.

Abstract

Background: Autophagy is highly active in neuroepithelial cells of the developing neuroepithelium, and impairment of autophagy leads to neural tube defects. In this study, we have found that maternal diabetes suppresses autophagy that leads to neural tube defects and consequent cellular imbalance in the endoplasmic reticulum where critical events occur, leading to the induction of diabetic embryopathy. Because the mammalian target of rapamycin pathway suppresses autophagy, we hypothesized that 70 kDa ribosomal protein S6 kinase 1 (p70S6K1), a major downstream effector of mammalian target of rapamycin, mediates the inhibitory effect of maternal diabetes on autophagy in the developing neuroepithelium.

Objective: We investigated whether p70S6K1 mediates the inhibitory effect of maternal diabetes on autophagy during neurulation. We also examined whether p70S6K1 deficiency restores autophagy and therefore relieves endoplasmic reticulum stress and inhibits maternal diabetes-induced apoptosis, which leads to reduction in neural tube defect incidence in diabetic embryopathy.

Study design: Female p70S6K1 heterogeneous knockout (p70S6K1+/-) mice were bred with male p70S6K1 heterogeneous knockout (p70S6K1+/-) mice to generate wild-type (WT), p70S6K1+/- and p70S6K1 knockout (p70S6K1-/-) embryos. Embryos at embryonic day 8.5 were harvested for the assessment of indices of autophagy, endoplasmic reticulum stress, and apoptosis. Neural tube defect incidence in embryos was determined at embryonic day 10.5. For in vitro studies, small interfering RNA knockdown of p70S6K1 in C17.2 mouse neural stem cells was used to determine the effect of p70S6K1 deficiency on autophagy impairment and endoplasmic reticulum stress under high glucose conditions.

Results: Knockout of the Rps6kb1 gene, which encodes for p70S6K1, ameliorated maternal diabetes-induced NTDs and restored autophagosome formation in neuroepithelial cells suppressed by maternal diabetes. Maternal diabetes-suppressed conversion of LC3-I (microtubule-associated protein 1A/1B-light chain 3) to LC3-II, an index of autophagic activity, in neurulation stage embryos was abrogated in the absence of p70S6K1. p70S6K1 knockdown in neural stem cells also restored autophagosome formation and the conversion of LC3-I to LC3-II. The activation of the major unfolded protein response, indicated by phosphorylation of inositol-requiring enzyme 1 alpha, and protein kinase R-like endoplasmic reticulum kinase, and eukaryotic translation initiation factor 2α, and the increase of the endoplasmic reticulum stress marker, C/EBP homologous protein, were induced by maternal diabetes in vivo and high glucose in vitro. Unfolded protein response and endoplasmic reticulum stress induced by maternal diabetes or high glucose were reduced by Rps6kb1 deletion or p70S6K1 knockdown, respectively. Rps6kb1 knockout blocked maternal diabetes-induced caspase cleavage and neuroepithelial cell apoptosis. The superoxide dismutase mimetic Tempol abolished high glucose-induced p70S6K1 activation.

Conclusion: The study revealed the critical involvement of p70S6K1 in the pathogenesis of diabetic embryopathy.

Keywords: apoptosis; autophagy; diabetic embryopathy; endoplasmic reticulum stress; maternal diabetes; neural tube defects; p70S6K1.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Antioxidants / pharmacology
  • Apoptosis / drug effects
  • Apoptosis / genetics
  • Autophagosomes / drug effects
  • Autophagosomes / metabolism
  • Autophagy / genetics*
  • Blood Glucose / metabolism
  • Cyclic N-Oxides / pharmacology
  • Diabetes Mellitus, Experimental / complications
  • Diabetes Mellitus, Experimental / metabolism
  • Diabetes Mellitus, Type 1 / complications
  • Diabetes Mellitus, Type 1 / metabolism
  • Endoplasmic Reticulum Stress / drug effects
  • Endoplasmic Reticulum Stress / genetics*
  • Female
  • Fetal Diseases / etiology
  • Fetal Diseases / genetics*
  • Fetal Diseases / metabolism
  • Glucose / pharmacology
  • In Vitro Techniques
  • Mice
  • Mice, Knockout
  • Microtubule-Associated Proteins / metabolism
  • Neural Stem Cells / drug effects
  • Neural Stem Cells / metabolism*
  • Neural Tube Defects / embryology
  • Neural Tube Defects / genetics*
  • Neural Tube Defects / metabolism
  • Neuroepithelial Cells / drug effects
  • Neuroepithelial Cells / metabolism
  • Neurulation / genetics
  • Oxidative Stress
  • Pregnancy
  • Pregnancy in Diabetics / genetics*
  • Pregnancy in Diabetics / metabolism
  • Ribosomal Protein S6 Kinases, 70-kDa / genetics*
  • Spin Labels
  • Unfolded Protein Response / drug effects
  • Unfolded Protein Response / genetics*

Substances

  • Antioxidants
  • Blood Glucose
  • Cyclic N-Oxides
  • Map1lc3b protein, mouse
  • Microtubule-Associated Proteins
  • Spin Labels
  • Ribosomal Protein S6 Kinases, 70-kDa
  • ribosomal protein S6 kinase, 70kD, polypeptide 1
  • Glucose
  • tempol