Hostname: page-component-76fb5796d-zzh7m Total loading time: 0 Render date: 2024-04-26T04:01:44.977Z Has data issue: false hasContentIssue false

Adipose tissue uncoupling protein 1 levels and function are increased in a mouse model of developmental obesity induced by maternal exposure to high-fat diet

Published online by Cambridge University Press:  17 May 2018

E. Bytautiene Prewit*
Affiliation:
Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
C. Porter
Affiliation:
Department of Surgery, The University of Texas Medical Branch at Galveston, Shriners Hospitals for Children, Galveston, TX, USA
M. La Rosa
Affiliation:
Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
N. Bhattarai
Affiliation:
Department of Surgery, The University of Texas Medical Branch at Galveston, Shriners Hospitals for Children, Galveston, TX, USA
H. Yin
Affiliation:
Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
P. Gamble
Affiliation:
Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
T. Kechichian
Affiliation:
Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
L. S. Sidossis
Affiliation:
Department of Surgery, The University of Texas Medical Branch at Galveston, Shriners Hospitals for Children, Galveston, TX, USA Department of Kinesiology and Health Department of Medicine, Rutgers University, New Brunswick, NJ, USA
*
*Address for correspondence: E. Bytautiene Prewit, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX 77555-1062, USA. E-mail: egbytaut@utmb.edu

Abstract

With brown adipose tissue (BAT) becoming a possible therapeutic target to counteract obesity, the prenatal environment could represent a critical window to modify BAT function and browning of white AT. We investigated if levels of uncoupling protein 1 (UCP1) and UCP1-mediated thermogenesis are altered in offspring exposed to prenatal obesity. Female CD-1 mice were fed a high-fat (HF) or standard-fat (SF) diet for 3 months before breeding. After weaning, all pups were placed on SF. UCP1 mRNA and protein levels were quantified using quantitative real-time PCR and Western blot analysis, respectively, in brown (BAT), subcutaneous (SAT) and visceral (VAT) adipose tissues at 6 months of age. Total and UCP1-dependent mitochondrial respiration were determined by high-resolution respirometry. A Student’s t-test and Mann–Whitney test were used (significance: P<0.05). UCP1 mRNA levels were not different between the HF and SF offspring. UCP1 protein levels, total mitochondrial respiration and UCP1-dependent respiration were significantly higher in BAT from HF males (P=0.02, P=0.04, P=0.005, respectively) and females (P=0.01, P=0.04, P=0.02, respectively). In SAT, the UCP1 protein was significantly lower in HF females (P=0.03), and the UCP1-dependent thermogenesis was significantly lower from HF males (P=0.04). In VAT, UCP1 protein levels and UCP1-dependent respiration were significantly lower only in HF females (P=0.03, P=0.04, respectively). There were no differences in total respiration in SAT and VAT. Prenatal exposure to maternal obesity leads to significant increases in UCP1 levels and function in BAT in offspring with little impact on UCP1 levels and function in SAT and VAT.

Type
Original Article
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2018 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

1. Writing, G, Roger, VL, Go, AS, et al. Executive summary: heart disease and stroke statistics – 2011 update. Circulation. 2011; 123, 459463.Google Scholar
2. Fryar, CD, Hirsch, R, Eberhardt, MS, Yoon, SS, Wright, JD. Hypertension, high serum total cholesterol, and diabetes: racial and ethnic prevalence differences in U.S. adults, 1999-2006. NCHS Data Brief. 2010; 36, 18.Google Scholar
3. American Heart Association. Statistical fact sheet. Women and Cardiovascular Diseases: Statistics, 2006. Retrieved 1 May 2006 from http://wwwamericanheartorg/downloadable/heart/1136818052118Females06pdf.Google Scholar
4. Thom, T, Haase, N, Rosamond, W, et al. Heart disease and stroke statistics – 2006 update: a report from the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation. 2006; 113, e85e151.Google Scholar
5. Huda, SS, Brodie, LE, Sattar, N. Obesity in pregnancy: prevalence and metabolic consequences. Semin Fetal Neonatal Med. 2010; 15, 7076.Google Scholar
6. Galtier-Dereure, F, Boegner, C, Bringer, J. Obesity and pregnancy: complications and cost. American J Clin Nutr. 2000; 71(5 Suppl.), 1242S1248S.Google Scholar
7. Kim, SY, Dietz, PM, England, L, Morrow, B, Callaghan, WM. Trends in pre-pregnancy obesity in nine states, 1993-2003. Obesity. 2007; 15, 986993.Google Scholar
8. Lu, GC, Rouse, DJ, DuBard, M, et al. The effect of the increasing prevalence of maternal obesity on perinatal morbidity. Am J Obstet Gynecol. 2001; 185, 845849.Google Scholar
9. Catalano, PM, Ehrenberg, HM. The short- and long-term implications of maternal obesity on the mother and her offspring. BJOG. 2006; 113, 11261133.Google Scholar
10. Whitaker, RC. Predicting preschooler obesity at birth: the role of maternal obesity in early pregnancy. Pediatrics. 2004; 114, e29e36.Google Scholar
11. Boney, CM, Verma, A, Tucker, R, Vohr, BR. Metabolic syndrome in childhood: association with birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics. 2005; 115, e290e296.Google Scholar
12. Koupil, I, Toivanen, P. Social and early-life determinants of overweight and obesity in 18-year-old Swedish men. Int J Obes. 2008; 32, 7381.Google Scholar
13. Mingrone, G, Manco, M, Valera Mora, ME, et al. Influence of maternal obesity on insulin sensitivity and secretion in offspring. Diabetes Care. 2008; 31, 18721876.Google Scholar
14. Desai, M, Jellyman, JK, Han, G, et al. Maternal obesity and high-fat diet program offspring metabolic syndrome. Am J Obstet Gynecol. 2014; 211, 237.e231237.e237.Google Scholar
15. Forsen, T, Eriksson, JG, Tuomilehto, J, et al. Mother’s weight in pregnancy and coronary heart disease in a cohort of Finnish men: follow up study. BMJ. 1997; 315, 837840.Google Scholar
16. Khan, IY, Dekou, V, Douglas, G, et al. A high-fat diet during rat pregnancy or suckling induces cardiovascular dysfunction in adult offspring. Am J Physiol Regul Integr Comp Physiol. 2005; 288, R127R133.Google Scholar
17. Khan, IY, Taylor, PD, Dekou, V, et al. Gender-linked hypertension in offspring of lard-fed pregnant rats. Hypertension. 2003; 41, 168175.Google Scholar
18. Samuelsson, AM, Matthews, PA, Argenton, M, et al. Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: a novel murine model of developmental programming. Hypertension. 2008; 51, 383392.Google Scholar
19. Oben, JA, Mouralidarane, A, Samuelsson, AM, et al. Maternal obesity during pregnancy and lactation programs the development of offspring non-alcoholic fatty liver disease in mice. J Hepatol. 2010; 52, 913920.Google Scholar
20. Oben, JA, Patel, T, Mouralidarane, A, et al. Maternal obesity programmes offspring development of non-alcoholic fatty pancreas disease. Biochem Biophys Res Commun. 2010; 394, 2428.Google Scholar
21. Suter, MA, Ma, J, Vuguin, PM, et al. In utero exposure to a maternal high-fat diet alters the epigenetic histone code in a murine model. Am J Obstet Gynecol. 2014; 210, 463463.Google Scholar
22. Persichetti, A, Sciuto, R, Rea, S, et al. Prevalence, mass, and glucose-uptake activity of 18F-FDG-detected brown adipose tissue in humans living in a temperate zone of Italy. PLoS One. 2013; 8, e63391.Google Scholar
23. Lo Kinyui, A, Sun, L. Turning WAT into BAT: a review on regulators controlling the browning of white adipocytes. Biosci Rep. 2013; 33, e00065.Google Scholar
24. Fenzl, A, Kiefer, FW. Brown adipose tissue and thermogenesis. Horm Mol Biol Clin Investig. 2014; 19, 2537.Google Scholar
25. Porter, C, Herndon, DN, Bhattarai, N, et al. Severe burn injury induces thermogenically functional mitochondria in murine white adipose tissue. Shock. 2015; 44, 258264.Google Scholar
26. Bytautiene, E, Tamayo, E, Kechichian, T, et al. Prepregnancy obesity and sFlt1-induced preeclampsia in mice: developmental programming model of metabolic syndrome. Am J Obstet Gynecol. 2011; 204, 398.e391398.e398.Google Scholar
27. Porter, C, Hurren, NM, Cotter, MV, et al. Mitochondrial respiratory capacity and coupling control decline with age in human skeletal muscle. Am J Physiol Endocrinol Metab. 2015; 309, E224E232.Google Scholar
28. Cannon, B, Nedergaard, J. Respiratory and thermogenic capacities of cells and mitochondria from brown and white adipose tissue. Methods Mol Biol. 2001; 155, 295303.Google Scholar
29. Cannon, B, Nedergaard, J. Nonshivering thermogenesis and its adequate measurement in metabolic studies. J Exp Biol. 2010; 214, 242253.Google Scholar
30. Cannon, B, Nedergaard, J. Studies of thermogenesis and mitochondrial function in adipose tissues. Methods Mol Biol. 2008; 456, 109121.Google Scholar
31. Cannon, B, Nedergaard, J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004; 84, 277359.Google Scholar
32. Matthias, A, Ohlson, KB, Fredriksson, JM, et al. Thermogenic responses in brown fat cells are fully UCP1-dependent. UCP2 or UCP3 do not substitute for UCP1 in adrenergically or fatty acid-induced thermogenesis. J Biol Chem. 2000; 275, 2507325081.Google Scholar
33. Nedergaard, J, Cannon, B. UCP1 mRNA does not produce heat. Biochim Biophys Acta Mol Cell Biol Lipids. 2013; 1831, 943949.Google Scholar
34. Shabalina, IG, Ost, M, Petrovic, N, et al. Uncoupling protein-1 is not leaky. Biochim Biophys Acta. 2010; 1797, 773784.Google Scholar
35. Shabalina, IG, Petrovic, N, de Jong, JM, et al. UCP1 in brite/beige adipose tissue mitochondria is functionally thermogenic. Cell Rep. 2013; 5, 11961203.Google Scholar
36. Elahi, MM, Cagampang, FR, Mukhtar, D, et al. Long-term maternal high-fat feeding from weaning through pregnancy and lactation predisposes offspring to hypertension, raised plasma lipids and fatty liver in mice. Br J Nutr. 2009; 102, 514519.Google Scholar
37. Watkins, AJ, Lucas, ES, Wilkins, A, Cagampang, FRA, Fleming, TP. Maternal periconceptional and gestational low protein diet affects mouse offspring growth, cardiovascular and adipose phenotype at 1 year of age. PLoS One. 2011; 6, e28745.Google Scholar
38. Ojha, S, Robinson, L, Yazdani, M, Symonds, ME, Budge, H. Brown adipose tissue genes in pericardial adipose tissue of newborn sheep are downregulated by maternal nutrient restriction in late gestation. Pediatr Res. 2013; 74, 246251.Google Scholar
39. Felipe, A, Villarroya, F, Mampel, T. Effects of maternal hypocaloric diet feeding on neonatal rat brown adipose tissue. Biol Neonate. 1988; 53, 105112.Google Scholar
40. Priego, T, Sanchez, J, Garcia, A, Palou, A, Pico, C. Maternal dietary fat affects milk fatty acid profile and impacts on weight gain and thermogenic capacity of suckling rats. Lipids. 2013; 48, 481495.Google Scholar
41. Myers, DA, Hanson, K, Mlynarczyk, M, Kaushal, KM, Ducsay, CA. Long-term hypoxia modulates expression of key genes regulating adipose function in the late-gestation ovine fetus. Am J Physiol Regul Integr Comp Physiol. 2008; 294, R1312R1318.Google Scholar
42. de Almeida, DL, Fabricio, GS, Trombini, AB, et al. Early overfeed-induced obesity leads to brown adipose tissue hypoactivity in rats. Cell Physiol Biochem. 2013; 32, 16211630.Google Scholar
43. Xiao, XQ, Williams, SM, Grayson, BE, et al. Excess weight gain during the early postnatal period is associated with permanent reprogramming of brown adipose tissue adaptive thermogenesis. Endocrinology. 2007; 148, 41504159.Google Scholar
44. Carey Satterfield, M, Dunlap, K, Keisler, D, Bazer, F, Wu, G. Arginine nutrition and fetal brown adipose tissue development in diet-induced obese sheep. Amino Acids. 2012; 43, 15931603.Google Scholar
45. Maurer, AD, Reimer, RA. Maternal consumption of high-prebiotic fibre or -protein diets during pregnancy and lactation differentially influences satiety hormones and expression of genes involved in glucose and lipid metabolism in offspring in rats. Br J Nutr. 2011; 105, 329338.Google Scholar
46. Arias, N, Aguirre, L, Fernandez-Quintela, A, et al. MicroRNAs involved in the browning process of adipocytes. J Physiol Biochem. 2015; 72, 509521.Google Scholar
47. Guller, I, McNaughton, S, Crowley, T, et al. Comparative analysis of microRNA expression in mouse and human brown adipose tissue. BMC Genomics. 2015; 16, 111.Google Scholar
48. Quevedo, S, Roca, P, Pico, C, Palou, A. Sex-associated differences in cold-induced UCP1 synthesis in rodent brown adipose tissue. Pflugers Arch. 1998; 436, 689695.Google Scholar