4.7 Article

Mitochondrial Transfer Improves Cardiomyocyte Bioenergetics and Viability in Male Rats Exposed to Pregestational Diabetes

Journal

Publisher

MDPI
DOI: 10.3390/ijms22052382

Keywords

mitochondria; diabetic pregnancy; developmentally programmed heart disease; mitochondrial transfer

Funding

  1. National Institute of Child Health and Human Development (NIH-NICHD) [K08 HD078504]
  2. Sanford Center for Pediatric Research [NIH-NIGMS CoBRE 2P20GM103620-06]
  3. Sanford Center for Cancer Biology Research [NIH-NIGMS CoBRE P20GM103548]
  4. Wesley W. Parke Research Award Endowment

Ask authors/readers for more resources

Offspring born to diabetic or obese mothers have a higher lifetime risk of heart disease. Research has shown that exposure to late-gestational diabetes mellitus (LGDM) or maternal high-fat (HF) diet can lead to mitochondrial dysfunction, impaired cardiomyocyte bioenergetics, and cardiac dysfunction in rat offspring. Findings suggest that mitochondrial dysfunction may play a central role in neonatal cardiomyopathy, with sex-specific differences observed in mitochondria-mediated mechanisms of heart disease development. Therapeutic mitochondrial transfer may have potential limitations, such as inconsistent effects in male and female offspring.
Offspring born to diabetic or obese mothers have a higher lifetime risk of heart disease. Previously, we found that rat offspring exposed to late-gestational diabetes mellitus (LGDM) and maternal high-fat (HF) diet develop mitochondrial dysfunction, impaired cardiomyocyte bioenergetics, and cardiac dysfunction at birth and again during aging. Here, we compared echocardiography, cardiomyocyte bioenergetics, oxidative damage, and mitochondria-mediated cell death among control, pregestational diabetes mellitus (PGDM)-exposed, HF-diet-exposed, and combination-exposed newborn offspring. We hypothesized that PGDM exposure, similar to LGDM, causes mitochondrial dysfunction to play a central, pathogenic role in neonatal cardiomyopathy. We found that PGDM-exposed offspring, similar to LGDM-exposed offspring, have cardiac dysfunction at birth, but their isolated cardiomyocytes have seemingly less bioenergetics impairment. This finding was due to confounding by impaired viability related to poorer ATP generation, more lipid peroxidation, and faster apoptosis under metabolic stress. To mechanistically isolate and test the role of mitochondria, we transferred mitochondria from normal rat myocardium to control and exposed neonatal rat cardiomyocytes. As expected, transfer provides a respiratory boost to cardiomyocytes from all groups. They also reduce apoptosis in PGDM-exposed males, but not in females. Findings highlight sex-specific differences in mitochondria-mediated mechanisms of developmentally programmed heart disease and underscore potential caveats of therapeutic mitochondrial transfer.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available