Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Low birth weight is associated with adiposity, impaired skeletal muscle energetics and weight loss resistance in mice

Abstract

Background:

In utero undernutrition is associated with obesity and insulin resistance, although its effects on skeletal muscle remain poorly defined. Therefore, in the current study we explored the effects of in utero food restriction on muscle energy metabolism in mice.

Methods:

We used an experimental mouse model system of maternal undernutrition during late pregnancy to examine offspring from undernourished dams (U) and control offspring from ad libitum-fed dams (C). Weight loss of 10-week-old offspring on a 4-week 40% calorie-restricted diet was also followed. Experimental approaches included bioenergetic analyses in isolated mitochondria, intact (permeabilized) muscle and at the whole body level.

Results:

U have increased adiposity and decreased glucose tolerance compared to C. Strikingly, when U are put on a 40% calorie-restricted diet they lose half as much weight as calorie-restricted controls. Mitochondria from muscle overall from U had decreased coupled (state 3) and uncoupled (state 4) respiration and increased maximal respiration compared to C. Mitochondrial yield was lower in U than C. In permeabilized fiber preparations from mixed fiber-type muscle, U had decreased mitochondrial content and decreased adenylate-free leak respiration, fatty acid oxidative capacity and state 3 respiratory capacity through complex I. Fiber maximal oxidative phosphorylation capacity did not differ between U and C but was decreased with calorie restriction.

Conclusions:

Our results reveal that in utero undernutrition alters metabolic physiology through a profound effect on skeletal muscle energetics and blunts response to a hypocaloric diet in adulthood. We propose that mitochondrial dysfunction links undernutrition in utero with metabolic disease in adulthood.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

References

  1. Barker DJ, Osmond C, Golding J, Kuh D, Wadsworth ME . Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ 1989; 298: 564–567.

    Article  CAS  Google Scholar 

  2. Barker DJ, Hales CN, Fall CH, Osmond C, Phipps K, Clark PM . Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): relation to reduced fetal growth. Diabetologia 1993; 36: 62–67.

    Article  CAS  Google Scholar 

  3. Gluckman PD, Hanson MA, Cooper C, Thornburg KL . Effect of in utero and early-life conditions on adult health and disease. N Engl J Med 2008; 359: 61–73.

    Article  CAS  Google Scholar 

  4. Jimenez-Chillaron JC, Hernandez-Valencia M, Reamer C, Fisher S, Joszi A, Hirshman M et al. Beta-cell secretory dysfunction in the pathogenesis of low birth weight-associated diabetes: a murine model. Diabetes 2005; 54: 702–711.

    Article  CAS  Google Scholar 

  5. Isganaitis E, Jimenez-Chillaron J, Woo M, Chow A, DeCoste J, Vokes M et al. Accelerated postnatal growth increases lipogenic gene expression and adipocyte size in low-birth weight mice. Diabetes 2009; 58: 1192–1200.

    Article  CAS  Google Scholar 

  6. Tanner CJ, Barakat HA, Dohm GL, Pories WJ, MacDonald KG, Cunningham PR et al. Muscle fiber type is associated with obesity and weight loss. Am J Physiol Endocrinol Metab 2002; 282: E1191–E1196.

    Article  CAS  Google Scholar 

  7. Handschin C . Regulation of skeletal muscle cell plasticity by the peroxisome proliferator-activated receptor gamma coactivator 1alpha. J Recept Signal Transduct Res 2010; 30: 376–384.

    Article  CAS  Google Scholar 

  8. Fitts RH, Trappe SW, Costill DL, Gallagher PM, Creer AC, Colloton PA et al. Prolonged space flight-induced alterations in the structure and function of human skeletal muscle fibres. J Physiol 2010; 588: 3567–3592.

    Article  CAS  Google Scholar 

  9. He J, Watkins S, Kelley DE . Skeletal muscle lipid content and oxidative enzyme activity in relation to muscle fiber type in type 2 diabetes and obesity. Diabetes 2001; 50: 817–823.

    Article  CAS  Google Scholar 

  10. Oberbach A, Bossenz Y, Lehmann S, Niebauer J, Adams V, Paschke R et al. Altered fiber distribution and fiber-specific glycolytic and oxidative enzyme activity in skeletal muscle of patients with type 2 diabetes. Diabetes Care 2006; 29: 895–900.

    Article  CAS  Google Scholar 

  11. Jensen CB, Martin-Gronert MS, Storgaard H, Madsbad S, Vaag A, Ozanne SE . Altered PI3-kinase/Akt signalling in skeletal muscle of young men with low birth weight. PLoS One 2008; 3: e3738.

    Article  Google Scholar 

  12. Jensen CB, Storgaard H, Madsbad S, Richter EA, Vaag AA . Altered skeletal muscle fiber composition and size precede whole-body insulin resistance in young men with low birth weight. J Clin Endocrinol Metab 2007; 92: 1530–1534.

    Article  CAS  Google Scholar 

  13. Kensara OA, Wootton SA, Phillips DI, Patel M, Jackson AA, Elia M . Fetal programming of body composition: relation between birth weight and body composition measured with dual-energy X-ray absorptiometry and anthropometric methods in older Englishmen. Am J Clin Nutr 2005; 82: 980–987.

    Article  CAS  Google Scholar 

  14. Luquet S, Lopez-Soriano J, Holst D, Fredenrich A, Melki J, Rassoulzadegan M et al. Peroxisome proliferator-activated receptor delta controls muscle development and oxidative capability. FASEB J 2003; 17: 2299–2301.

    Article  CAS  Google Scholar 

  15. Fluck M, Hoppeler H . Molecular basis of skeletal muscle plasticity—from gene to form and function. Rev Physiol Biochem Pharmacol 2003; 146: 159–216.

    Article  CAS  Google Scholar 

  16. Park HK, Jin CJ, Cho YM, Park DJ, Shin CS, Park KS et al. Changes of mitochondrial DNA content in the male offspring of protein-malnourished rats. Ann N Y Acad Sci 2004; 1011: 205–216.

    Article  CAS  Google Scholar 

  17. Selak MA, Storey BT, Peterside I, Simmons RA . Impaired oxidative phosphorylation in skeletal muscle of intrauterine growth-retarded rats. Am J Physiol Endocrinol Metab 2003; 285: E130–E137.

    Article  CAS  Google Scholar 

  18. Boily G, Seifert EL, Bevilacqua L, He XH, Sabourin G, Estey C et al. SirT1 regulates energy metabolism and response to caloric restriction in mice. PLoS One 2008; 3: e1759.

    Article  Google Scholar 

  19. Du H, Dardzinski BJ, O'Brien KJ, Donnelly LF . MRI of fat distribution in a mouse model of lysosomal acid lipase deficiency. AJR Am J Roentgenol 2005; 184: 658–662.

    Article  Google Scholar 

  20. Estey C, Seifert EL, Aguer C, Moffat C, Harper ME . Calorie restriction in mice overexpressing UCP3: evidence that prior mitochondrial uncoupling alters response. Exp Gerontol 2012; 47: 361–371.

    Article  CAS  Google Scholar 

  21. Gerrits MF, Ghosh S, Kavaslar N, Hill B, Tour A, Seifert EL et al. Distinct skeletal muscle fiber characteristics and gene expression in diet-sensitive versus diet-resistant obesity. J Lipid Res 2010; 51: 2394–2404.

    Article  CAS  Google Scholar 

  22. Chappell JB, Perry SV . Biochemical and osmotic properties of skeletal muscle mitochondria. Nature 1954; 173: 1094–1095.

    Article  CAS  Google Scholar 

  23. Seifert EL, Estey C, Xuan JY, Harper ME . Electron transport chain-dependent and -independent mechanisms of mitochondrial H2O2 emission during long-chain fatty acid oxidation. J Biol Chem 2010; 285: 5748–5758.

    Article  CAS  Google Scholar 

  24. Rogers GW, Brand MD, Petrosyan S, Ashok D, Elorza AA, Ferrick DA et al. High throughput microplate respiratory measurements using minimal quantities of isolated mitochondria. PLoS One 2011; 6: e21746.

    Article  CAS  Google Scholar 

  25. Mailloux RJ, Xuan JY, Beauchamp B, Jui L, Lou M, Harper ME . Glutaredoxin-2 is required to control proton leak through uncoupling protein-3. J Biol Chem 2013; 288: 8365–8379.

    Article  CAS  Google Scholar 

  26. Harper ME, Dent R, Monemdjou S, Bezaire V, Van Wyck L, Wells G et al. Decreased mitochondrial proton leak and reduced expression of uncoupling protein 3 in skeletal muscle of obese diet-resistant women. Diabetes 2002; 51: 2459–2466.

    Article  CAS  Google Scholar 

  27. Pette D, Staron RS . Myosin isoforms, muscle fiber types, and transitions. Microsc Res Tech 2000; 50: 500–509.

    Article  CAS  Google Scholar 

  28. Ghosh S, Dent R, Harper ME, Stuart J, McPherson R . Blood gene expression reveal pathway differences between diet-sensitive and resistant obese subjects prior to caloric restriction. Obesity (Silver Spring) 2011; 19: 457–463.

    Article  Google Scholar 

  29. Rolfe DF, Brown GC . Cellular energy utilization and molecular origin of standard metab olic rate in mammals. Physiol Rev 1997; 77: 731–758.

    Article  CAS  Google Scholar 

  30. Zurlo F, Larson K, Bogardus C, Ravussin E . Skeletal muscle metabolism is a major determinant of resting energy expenditure. J Clin Invest 1990; 86: 1423–1427.

    Article  CAS  Google Scholar 

  31. Hediger ML, Overpeck MD, Kuczmarski RJ, McGlynn A, Maurer KR, Davis WW . Muscularity and fatness of infants and young children born small- or large-for-gestational-age. Pediatrics 1998; 102: E60.

    Article  CAS  Google Scholar 

  32. Rasmussen EL, Malis C, Jensen CB, Jensen JE, Storgaard H, Poulsen P et al. Altered fat tissue distribution in young adult men who had low birth weight. Diabetes Care 2005; 28: 151–153.

    Article  Google Scholar 

  33. Wells JC, Chomtho S, Fewtrell MS . Programming of body composition by early growth and nutrition. Proc Nutr Soc 2007; 66: 423–434.

    Article  Google Scholar 

  34. Woo M, Isganaitis E, Cerletti M, Fitzpatrick C, Wagers AJ, Jimenez-Chillaron J et al. Early life nutrition modulates muscle stem cell number: implications for muscle mass and repair. Stem Cells Dev 2011; 20: 1763–1769.

    Article  CAS  Google Scholar 

  35. Jimenez-Chillaron JC, Hernandez-Valencia M, Lightner A, Faucette RR, Reamer C, Przybyla R et al. Reductions in caloric intake and early postnatal growth prevent glucose intolerance and obesity associated with low birthweight. Diabetologia 2006; 49: 1974–1984.

    Article  CAS  Google Scholar 

  36. Miles JL, Huber K, Thompson NM, Davison M, Breier BH . Moderate daily exercise activates metabolic flexibility to prevent prenatally induced obesity. Endocrinology 2009; 150: 179–186.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank Jian Xuan for technical assistance with animal work, Linda Jui for preparation and staining of histological slides, and Gabrielle Côté for assistance with counting muscle fibers. This research was supported through grants from Canadian Institutes of Health Research (MOP57810, M-EH; MOP258677, AB), National Institutes of Health (P20MD000175, DK088319; SG) and American Heart Association (AHA10SDG4230068; SG). Scholarships were also awarded from Natural Sciences and Engineering Research Council of Canada (Canada Graduate Scholarship -Doctoral, BB; -Master’s, AC).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to M-E Harper.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Author Contributions

BB and M-EH conceived and/or designed the work; BB, MD, GNK, AC and KR performed experiments; BB, SG, MD, AC, AB, KR, ECT, M-EP and M-EH analyzed or interpreted results; BB and M-EH wrote the manuscript. All authors revised the manuscript and approved the final version.

Supplementary Information accompanies this paper on International Journal of Obesity website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Beauchamp, B., Ghosh, S., Dysart, M. et al. Low birth weight is associated with adiposity, impaired skeletal muscle energetics and weight loss resistance in mice. Int J Obes 39, 702–711 (2015). https://doi.org/10.1038/ijo.2014.120

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ijo.2014.120

Search

Quick links