Hostname: page-component-8448b6f56d-c4f8m Total loading time: 0 Render date: 2024-04-23T13:33:10.300Z Has data issue: false hasContentIssue false

Maternal malnutrition and placental insufficiency induce global downregulation of gene expression in fetal kidneys

Published online by Cambridge University Press:  10 December 2010

O. Denisenko*
Affiliation:
Department of Medicine, University of Washington, Seattle, WA, USA
B. Lin
Affiliation:
Division of Nephrology, Oregon Health & Science University, Portland, OR, USA Research Service, Portland VA Medical Center, Portland, OR, USA
S. Louey
Affiliation:
Heart Research Center, Oregon Health & Science University, Portland, OR, USA Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, OR, USA
K. Thornburg
Affiliation:
Heart Research Center, Oregon Health & Science University, Portland, OR, USA Division of Cardiovascular Medicine, Oregon Health & Science University, Portland, OR, USA
K. Bomsztyk
Affiliation:
Department of Medicine, University of Washington, Seattle, WA, USA
S. Bagby
Affiliation:
Heart Research Center, Oregon Health & Science University, Portland, OR, USA Division of Nephrology, Oregon Health & Science University, Portland, OR, USA Research Service, Portland VA Medical Center, Portland, OR, USA
*
*Address for correspondence: O. Denisenko, Department of Medicine, University of Washington, 815 Mercer Street, Room 244, Seattle, WA 98109, USA. (Email odenis@uw.edu)

Abstract

Malnutrition during pregnancy causes intrauterine growth restriction and long-term changes in the offspring's physiology and metabolism. To explore molecular mechanisms by which the intrauterine environment conveys programming in fetal kidneys, an organ known to undergo substantial changes in many animal models of late gestational undernutrition, we used a microswine model of maternal protein restriction (MPR) in which sows were exposed to isocaloric low protein (LP) diet during late gestation/early lactation to encompass the bulk of nephrogenesis. To define general v. model-specific effects, we also used a sheep model of placental insufficiency. In kidneys from near-term fetal and neonatal microswine LP offspring, per cell levels of total RNA, poly(A)+ mRNA and transcripts of several randomly chosen housekeeping genes were significantly reduced compared to controls. Microarray analysis revealed only a few MPR-resistant genes that escape such downregulation. The ratio of histone modifications H3K4m3/H3K9m3 (active/silenced) was reduced at promoters of downregulated but not MPR-resistant genes suggesting that transcriptional suppression is the point of control. In juvenile offspring, on a normal diet from weaning, cellular RNA levels and histone mark patterns were recovered to near control levels, indicating that global repression of transcription is dependent on ongoing MPR. Importantly, cellular RNA content was also reduced in ovine fetal kidneys during placental insufficiency. These studies show that global repression of transcription may be a universal consequence of a poor intrauterine environment that contributes to fetal restriction.

Type
Original Articles
Copyright
Copyright © Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2010

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. McCance, RA, Widdowson, EM. Nutrition and growth. Proc R Soc Lond B Biol Sci. 1962; 156, 326337.Google Scholar
2. McCance, RA, Widdowson, EM. The determinants of growth and form. Proc R Soc Lond B Biol Sci. 1974; 185, 117.Google ScholarPubMed
3. Barker, DJ, Bagby, SP, Hanson, MA. Mechanisms of disease: in utero programming in the pathogenesis of hypertension. Nat Clin Pract Nephrol. 2006; 2, 700707.CrossRefGoogle ScholarPubMed
4. Barker, DJ, Clark, PM. Fetal undernutrition and disease in later life. Rev Reprod. 1997; 2, 105112.CrossRefGoogle ScholarPubMed
5. Desai, M, Hales, CN. Role of fetal and infant growth in programming metabolism in later life. Biol Rev Camb Philos Soc. 1997; 72, 329348.CrossRefGoogle ScholarPubMed
6. Gillman, MW, Barker, D, Bier, D, et al. Meeting report on the 3rd international congress on developmental origins of health and disease (DOHaD). Pediatr Res. 2007; 61, 625629.CrossRefGoogle ScholarPubMed
7. Hales, CN, Barker, DJ. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia. 1992; 35, 595601.CrossRefGoogle ScholarPubMed
8. Harding, JE, Johnston, BM. Nutrition and fetal growth. Reprod Fertil Dev. 1995; 7, 539547.CrossRefGoogle ScholarPubMed
9. Wu, G, Bazer, FW, Cudd, TA, Meininger, CJ, Spencer, TE. Maternal nutrition and fetal development. J Nutr. 2004; 134, 21692172.Google ScholarPubMed
10. Atinmo, T, Pond, WG, Barnes, RH. Effect of maternal energy vs. protein restriction on growth and development of progeny in swine. J Anim Sci. 1974; 39, 703711.CrossRefGoogle ScholarPubMed
11. Pond, WG, Strachan, DN, Sinha, YN, et al. Effect of protein deprivation of swine during all or part of gestation on birth weight, postnatal growth rate and nucleic acid content of brain and muscle of progeny. J Nutr. 1969; 99, 6167.CrossRefGoogle ScholarPubMed
12. Mellor, DJ. Nutritional and placental determinants of foetal growth rate in sheep and consequences for the newborn lamb. Br Vet J. 1983; 139, 307324.CrossRefGoogle ScholarPubMed
13. Cock, ML, Camm, EJ, Louey, S, Joyce, BJ, Harding, R. Postnatal outcomes in term and preterm lambs following fetal growth restriction. Clin Exp Pharmacol Physiol. 2001; 28, 931937.CrossRefGoogle ScholarPubMed
14. Cock, ML, Harding, R. Renal and amniotic fluid responses to umbilicoplacental embolization for 20 days in fetal sheep. Am J Physiol. 1997; 273, R1094R1102.Google ScholarPubMed
15. Gagnon, R, Challis, J, Johnston, L, Fraher, L. Fetal endocrine responses to chronic placental embolization in the late-gestation ovine fetus. Am J Obstet Gynecol. 1994; 170, 929938.CrossRefGoogle ScholarPubMed
16. Louey, S, Cock, ML, Stevenson, KM, Harding, R. Placental insufficiency and fetal growth restriction lead to postnatal hypotension and altered postnatal growth in sheep. Pediatr Res. 2000; 48, 808814.CrossRefGoogle ScholarPubMed
17. Louey, S, Jonker, SS, Giraud, GD, Thornburg, KL. Placental insufficiency decreases cell cycle activity and terminal maturation in fetal sheep cardiomyocytes. J Physiol. 2007; 580, 639648.CrossRefGoogle ScholarPubMed
18. MacLennan, NK, James, SJ, Melnyk, S, et al. Uteroplacental insufficiency alters DNA methylation, one-carbon metabolism, and histone acetylation in IUGR rats. Physiol Genomics. 2004; 18, 4350.CrossRefGoogle ScholarPubMed
19. Morrison, JL, Botting, KJ, Dyer, JL, et al. Restriction of placental function alters heart development in the sheep fetus. Am J Physiol Regul Integr Comp Physiol. 2007; 293, R306R313.CrossRefGoogle ScholarPubMed
20. Murotsuki, J, Challis, JR, Han, VK, Fraher, LJ, Gagnon, R. Chronic fetal placental embolization and hypoxemia cause hypertension and myocardial hypertrophy in fetal sheep. Am J Physiol. 1997; 272, R201R207.Google ScholarPubMed
21. Chang, JH, Rutledge, JC, Stoops, D, Abbe, R. Hypobaric hypoxia-induced intrauterine growth retardation. Biol Neonate. 1984; 46, 1013.CrossRefGoogle ScholarPubMed
22. Bell, AW, Hay, WW Jr, Ehrhardt, RA. Placental transport of nutrients and its implications for fetal growth. J Reprod Fertil Suppl. 1999; 54, 401410.Google ScholarPubMed
23. Marsal, K. Intrauterine growth restriction. Curr Opin Obstet Gynecol. 2002; 14, 127135.CrossRefGoogle ScholarPubMed
24. McMillen, IC, Robinson, JS. Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol Rev. 2005; 85, 571633.CrossRefGoogle ScholarPubMed
25. Jaenisch, R, Bird, A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003; 33 (Suppl.), 245254.CrossRefGoogle ScholarPubMed
26. Aagaard-Tillery, K, Grove, K, Bishop, J, et al. Developmental origins of disease and determinants of chromatin structure: maternal diet modifies the primate fetal epigenome. J Mol Endocrinol. 2008; 41, 91102.CrossRefGoogle ScholarPubMed
27. Waterland, RA, Jirtle, RL. Early nutrition, epigenetic changes at transposons and imprinted genes, and enhanced susceptibility to adult chronic diseases. Nutrition. 2004; 20, 6368.CrossRefGoogle ScholarPubMed
28. Tobi, EW, Lumey, LH, Talens, RP, et al. DNA methylation differences after exposure to prenatal famine are common and timing- and sex-specific. Hum Mol Genet. 2009; 18, 40464053.CrossRefGoogle ScholarPubMed
29. Pham, TD, MacLennan, NK, Chiu, CT, et al. Uteroplacental insufficiency increases apoptosis and alters p53 gene methylation in the full-term IUGR rat kidney. Am J Physiol Regul Integr Comp Physiol. 2003; 285, R962R970.CrossRefGoogle ScholarPubMed
30. Simmons, RA. Developmental origins of diabetes: the role of epigenetic mechanisms. Curr Opin Endocrinol Diabetes Obes. 2007; 14, 1316.CrossRefGoogle ScholarPubMed
31. Vickaryous, N, Whitelaw, E. The role of early embryonic environment on epigenotype and phenotype. Reprod Fertil Dev. 2005; 17, 335340.CrossRefGoogle ScholarPubMed
32. Bagby, SP, Kupfer, P, Lin, B, et al. Accelerated growth without prepubertal obesity in nutritionally programmed microswine offspring. Submitted. 2010.Google Scholar
33. Bagby, SP, Kupfer, P, Lin, B, et al. Perinatal maternal protein restriction in microswine programs enhanced vascular reactivity to AngII via EGF receptor transactivation in normotensive juvenile offspring. Submitted. 2010.Google Scholar
34. Economides, DL, Nicolaides, KH. Blood glucose and oxygen tension levels in small-for-gestational-age fetuses. Am J Obstet Gynecol. 1989; 160, 385389.CrossRefGoogle ScholarPubMed
35. Economides, DL, Nicolaides, KH, Campbell, S. Metabolic and endocrine findings in appropriate and small for gestational age fetuses. J Perinat Med. 1991; 19, 97105.CrossRefGoogle ScholarPubMed
36. Langford, K, Blum, W, Nicolaides, K, et al. The pathophysiology of the insulin-like growth factor axis in fetal growth failure: a basis for programming by undernutrition? Eur J Clin Invest. 1994; 24, 851856.CrossRefGoogle ScholarPubMed
37. Nicolaides, KH, Economides, DL, Soothill, PW. Blood gases, pH, and lactate in appropriate- and small-for-gestational-age fetuses. Am J Obstet Gynecol. 1989; 161, 9961001.CrossRefGoogle ScholarPubMed
38. Cheung, CY, Bogic, L, Gagnon, R, Harding, R, Brace, RA. Morphologic alterations in ovine placenta and fetal liver following induced severe placental insufficiency. J Soc Gynecol Investig. 2004; 11, 521528.CrossRefGoogle ScholarPubMed
39. Kanno, J, Aisaki, K, Igarashi, K, et al. “Per cell” normalization method for mRNA measurement by quantitative PCR and microarrays. BMC Genomics. 2006; 7, 64.CrossRefGoogle ScholarPubMed
40. Flanagin, S, Nelson, JD, Castner, DG, Denisenko, O, Bomsztyk, K. Microplate-based chromatin immunoprecipitation method, Matrix ChIP: a platform to study signaling of complex genomic events. Nucleic Acids Res. 2008; 36, e17.CrossRefGoogle ScholarPubMed
41. Friis, C. Postnatal development of the pig kidney: ultrastucure of the glomerulus and the proximal tubule. J Anat. 1980; 130, 513526.Google ScholarPubMed
42. Eriksson, J, Forsen, T, Tuomilehto, J, Osmond, C, Barker, D. Fetal and childhood growth and hypertension in adult life. Hypertension. 2000; 36, 790794.CrossRefGoogle ScholarPubMed
43. Nichols, SD, Boyne, MS, Thame, M, et al. Cold-induced elevation of forearm vascular resistance is inversely related to birth weight. J Hum Hypertens. 2005; 19, 309314.CrossRefGoogle ScholarPubMed
44. Orlando, V, Strutt, H, Paro, R. Analysis of chromatin structure by in vivo formaldehyde cross-linking. Methods. 1997; 11, 205214.CrossRefGoogle ScholarPubMed
45. Nelson, JD, Denisenko, O, Bomsztyk, K. Protocol for the fast chromatin immunoprecipitation (ChIP) method. Nat Protoc. 2006; 1, 179185.CrossRefGoogle ScholarPubMed
46. Nelson, JD, Flanagin, S, Kawata, Y, Denisenko, O, Bomsztyk, K. Transcription of laminin {gamma}1 chain gene in rat mesangial cells: constitutive and inducible RNA polymerase II recruitment and chromatin states. Am J Physiol Renal Physiol. 2008; 294, F525F533.CrossRefGoogle ScholarPubMed
47. Denisenko, O, Bomsztyk, K. Epistatic interaction between the K-homology domain protein HEK2 and SIR1 at HMR and telomeres in yeast. J Mol Biol. 2008; 375, 11781187.CrossRefGoogle ScholarPubMed
48. Barski, A, Cuddapah, S, Cui, K, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007; 129, 823837.CrossRefGoogle ScholarPubMed
49. Bagby, SP. Maternal nutrition, low nephron number, and hypertension in later life: pathways of nutritional programming. J Nutr. 2007; 137, 10661072.CrossRefGoogle ScholarPubMed
50. Ingelfinger, JR, Schnaper, HW. Renal endowment: developmental origins of adult disease. J Am Soc Nephrol. 2005; 16, 25332536.CrossRefGoogle ScholarPubMed
51. Coles, HS, Burne, JF, Raff, MC. Large-scale normal cell death in the developing rat kidney and its reduction by epidermal growth factor. Development. 1993; 118, 777784.CrossRefGoogle ScholarPubMed
52. Melamed, J, Darzynkiewicz, Z. RNA content and chromatin structure of CHO cells arrested in metaphase by colcemid. Cytometry. 1985; 6, 381385.CrossRefGoogle ScholarPubMed
53. Haugen, G, Hanson, M, Kiserud, T, et al. Fetal liver-sparing cardiovascular adaptations linked to mother's slimness and diet. Circ Res. 2005; 96, 1214.CrossRefGoogle ScholarPubMed
54. Kiserud, T, Kessler, J, Ebbing, C, Rasmussen, S. Ductus venosus shunting in growth-restricted fetuses and the effect of umbilical circulatory compromise. Ultrasound Obstet Gynecol. 2006; 28, 143149.CrossRefGoogle ScholarPubMed
55. Peeters, LL, Sheldon, RE, Jones, MD Jr, Makowski, EL, Meschia, G. Blood flow to fetal organs as a function of arterial oxygen content. Am J Obstet Gynecol. 1979; 135, 637646.CrossRefGoogle ScholarPubMed
56. Grummt, I, Smith, VA, Grummt, F. Amino acid starvation affects the initiation frequency of nucleolar RNA polymerase. Cell. 1976; 7, 439445.CrossRefGoogle ScholarPubMed
57. Kuwano, M, Endo, H, Ikehera, Y. Differences in RNA formation and polyribosome metabolism in serum-starved normal and transformed cells. Cancer Res. 1973; 33, 29652971.Google ScholarPubMed
58. Moss, T, Stefanovsky, VY. Promotion and regulation of ribosomal transcription in eukaryotes by RNA polymerase I. Prog Nucleic Acid Res Mol Biol. 1995; 50, 2566.CrossRefGoogle ScholarPubMed
59. Jenuwein, T, Allis, CD. Translating the histone code. Science. 2001; 293, 10741080.CrossRefGoogle ScholarPubMed
60. Guenther, MG, Levine, SS, Boyer, LA, Jaenisch, R, Young, RA. A chromatin landmark and transcription initiation at most promoters in human cells. Cell. 2007; 130, 7788.CrossRefGoogle ScholarPubMed
61. Kapranov, P, Cawley, SE, Drenkow, J, et al. Large-scale transcriptional activity in chromosomes 21 and 22. Science. 2002; 296, 916919.CrossRefGoogle Scholar
62. Robbins, ME, Stephens, LC, Johnston, DA, et al. A morphometric analysis of glomerular and tubular alterations following fast-neutron irradiation of the pig and monkey kidney. Int J Radiat Oncol Biol Phys. 1998; 41, 11491156.CrossRefGoogle ScholarPubMed
63. Tuchmann-Duplessis, H, Hiss, D, Mottot, G, Rosner, I. Embryotoxic and teratogenic effect of actinomycin D in the Syrian hamster. Toxicology. 1973; 1, 131133.CrossRefGoogle ScholarPubMed
64. Karnofsky, DA. Drugs as teratogens in animals and man. Annu Rev Pharmacol. 1965; 10, 447472.CrossRefGoogle Scholar
Supplementary material: File

Denisenko supplementary material

Table S1.doc

Download Denisenko supplementary material(File)
File 34.3 KB