Article Text

Download PDFPDF

Increased placental mitochondrial fusion in gestational diabetes mellitus: an adaptive mechanism to optimize feto-placental metabolic homeostasis?
  1. Joelcio Abbade1,2,
  2. Miira Marjuska Klemetti1,3,4,
  3. Abby Farrell1,
  4. Leonardo Ermini1,
  5. Taylor Gillmore1,5,
  6. Julien Sallais1,5,
  7. Andrea Tagliaferro1,
  8. Martin Post5,6,
  9. Isabella Caniggia1,4,5
  1. 1Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
  2. 2Departamento de Ginecologia e Obstetrícia Faculdade de Medicina de Botucatu, Sao Paulo, Brazil
  3. 3Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
  4. 4Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
  5. 5Department of Physiology and Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
  6. 6Hospital for Sick Children SickKids Learning Institute, Toronto, Ontario, Canada
  1. Correspondence to Dr Isabella Caniggia; caniggia{at}lunenfeld.ca

Abstract

Introduction Gestational diabetes mellitus (GDM), a common pregnancy disorder, increases the risk of fetal overgrowth and later metabolic morbidity in the offspring. The placenta likely mediates these sequelae, but the exact mechanisms remain elusive. Mitochondrial dynamics refers to the joining and division of these organelles, in attempts to maintain cellular homeostasis in stress conditions or alterations in oxygen and fuel availability. These remodeling processes are critical to optimize mitochondrial function, and their disturbances characterize diabetes and obesity.

Methods and results Herein we show that placental mitochondrial dynamics are tilted toward fusion in GDM, as evidenced by transmission electron microscopy and changes in the expression of key mechanochemical enzymes such as OPA1 and active phosphorylated DRP1. In vitro experiments using choriocarcinoma JEG-3 cells demonstrated that increased exposure to insulin, which typifies GDM, promotes mitochondrial fusion. As placental ceramide induces mitochondrial fission in pre-eclampsia, we also examined ceramide content in GDM and control placentae and observed a reduction in placental ceramide enrichment in GDM, likely due to an insulin-dependent increase in ceramide-degrading ASAH1 expression.

Conclusions Placental mitochondrial fusion is enhanced in GDM, possibly as a compensatory response to maternal and fetal metabolic derangements. Alterations in placental insulin exposure and sphingolipid metabolism are among potential contributing factors. Overall, our results suggest that GDM has profound impacts on placental mitochondrial dynamics and metabolism, with plausible implications for the short-term and long-term health of the offspring.

  • placenta
  • GDM
  • mitochondria
http://creativecommons.org/licenses/by-nc/4.0/

This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/.

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Footnotes

  • JA and MMK contributed equally.

  • Contributors JA carried out the cytotrophoblast transmission electron microscopy (TEM) analyses, western blot (WB) and half of immunofluorescence (IF) experiments. MMK performed half of IF experiments and participated in WBs together with AT and AF and analyzed clinical data. MMK, AF and JS carried out STB and endothelium TEM analyses. LE performed the studies on mitochondrial isolates and prepared samples for lipidomic analyses. TG prepared figure 1. MMK and IC wrote the manuscript, and all authors contributed toward data interpretation and critically revising the paper and accepted the final version. MMK and IC are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

  • Funding This work was supported by the Canadian Institutes for Health Research (MOP-133436 to IC), National Institutes of Health (1R01HD089660 to IC), Finnish Cultural Foundation (MMK) and Biomedicum Helsinki Research Foundation (MMK).

  • Competing interests None declared.

  • Patient consent for publication Not required.

  • Ethics approval The study protocol was approved by the MSH Research Ethics Board (REB number: 11–0287-E) and carried out in agreement with the Declaration of Helsinki.

  • Provenance and peer review Not commissioned; externally peer reviewed.

  • Data availability statement Data are available on reasonable request. The deidentified data generated and/or analyzed in this study are available from the corresponding author (caniggia@lunenfeld.ca) on reasonable request.