Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-19T01:38:39.162Z Has data issue: false hasContentIssue false

Early development and osteoporosis and bone health

Published online by Cambridge University Press:  08 December 2009

E. M. Dennison*
Affiliation:
Reader and Honorary Consultant in Rheumatology, MRC Epidemiology Resource Centre, Southampton General Hospital, University of Southampton, Southampton, England, UK
C. Cooper
Affiliation:
Professor of Rheumatology and Director, MRC Epidemiology Resource Centre, Southampton General Hospital, University of Southampton, Southampton, England, UK and Norman Collison Chair of Musculo-Skeletal Sciences, University of Oxford
Z. A. Cole
Affiliation:
ARC Clinical Research Fellow, MRC Epidemiology Resource Centre, Southampton General Hospital, University of Southampton, Southampton, England, UK
*
Address for correspondence: E. M. Dennison, Reader and Honorary Consultant in Rheumatology, MRC Epidemiology Resource Centre, Southampton General Hospital, University of Southampton, Southampton SO16 6YD, England, UK. (Email emd@mrc.soton.ac.uk)

Abstract

Osteoporosis is a skeletal disorder characterized by low bone mass and micro-architectural deterioration of bone tissue with a consequent increase in bone fragility and susceptibility to fracture. Evidence is now accumulating from human studies that programming of bone growth might be an important contributor to the later risk of osteoporotic fracture. Body weight in infancy is a determinant of adult bone mineral content, as well as of the basal levels of activity of the growth hormone/insulin-like growth factor-1 (GH/IGF-1) and hypothalamo-pituitary-adrenal (HPA) axes, and recent work has suggested a central role for vitamin D. Epidemiological studies have shown that maternal smoking and nutrition during pregnancy influence intrauterine skeletal mineralization. Childhood growth rates have been directly linked to the risk of hip fracture many decades later, and now evidence is emerging from experimental animal studies that support these observational data. Recent studies have also highlighted epigenetic phenomena as potential mechanisms underlying the findings from epidemiological studies.

Type
Reviews
Copyright
Copyright © Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2009

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. Consensus Development Conference. Prophylaxis and treatment of osteoporosis. Osteoporosis Int. 1991; 1, 114117.Google Scholar
2. Kovacs, CS. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism (ed. Favus MJ), 2003; pp. 6571. ASBMR, Washington, DC.Google Scholar
3. Gilsanz, V, Nelson, DA. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism (ed. Favus MJ), 2003; pp. 7179. ASBMR, Wahsington, DC.Google Scholar
4. Hui, SL, Slemenda, CW, Johnston, CC Jr. The contribution of bone loss to postmenopausal osteoporosis. Osteoporos. Int. 1990; 1, 3034.Google Scholar
5. Dennison, E, Cole, Z, Cooper, C. Diagnosis and epidemiology of osteoporosis. Curr Opin Rheumatol. 2005; 17, 456461.Google Scholar
6. Barker, DJP. Mothers, Babies and Diseases in Later Life, 1994. BMJ Publishing group, London, UK.Google Scholar
7. Hernandez, CJ, Beaupre, GS, Carter, DR. A theoretical analysis of the relative influences of peak BMD, age-related bone loss and menopause on the development of osteoporosis. Osteoporos Int. 2003; 14, 843847.Google Scholar
8. Barker, DJP. The fetal origins of adult disease. Proc R Soc Lond B. 1995; 262, 3743.Google Scholar
9. DeLise, AM, Fischer, L, Tuan, RS. Cellular interactions and signaling in cartilage development. Osteoarthritis Cartilage. 2000; 8, 309334.Google Scholar
10. Karaplis, AC, Luz, A, Glowacki, J, et al. Lethal skeletal dysplasia from targeted disruption of the parathyroid hormone-related peptide gene. Genes Dev. 1994; 8, 277289.Google Scholar
11. Bhaumick, B, Bala, RM. Differential effects of insulin-like growth factors I and II on growth, differentiation and glucoregulation in differentiating chondrocyte cells in culture. Acta Endocrinol (Copenh). 1991; 125, 201211.Google Scholar
12. Sylvia, VL, Del Toro, F, Hardin, RR, et al. Characterization of PGE(2) receptors (EP) and their role as mediators of 1alpha,25-(OH)(2)D(3) effects on growth zone chondrocytes. J Steroid Biochem Mol Biol. 2001; 78, 261274.Google Scholar
13. Quarto, R, Campanile, G, Cancedda, R, Dozin, B. Modulation of commitment, proliferation, and differentiation of chondrogenic cells in defined culture medium. Endocrinology. 1997; 138, 49664976.Google Scholar
14. Ducy, P. Cbfa1: a molecular switch in osteoblast biology. Dev Dyn. 2000; 219, 461471.Google Scholar
15. Schauberger, CW, Pitkin, RM. Maternal–perinatal calcium relationships. Obstet Gynecol. 1979; 53, 7476.Google Scholar
16. Forester, F, Daffos, F, Rainaut, M, Bruneau, M, Trivin, F. Blood chemistry of normal human fetuses at mid-trimester of pregnancy. Paed Res. 1987; 21, 579.Google Scholar
17. Ardawi, MS, Nasrat, HA, BA’Aqueel, HS. Calcium-regulating hormones and parathyroid hormone-related peptide in normal human pregnancy and postpartum: a longitudinal study. Eur J Endocrinol. 1997; 137, 402409.Google Scholar
18. Hosking, DJ. Calcium homeostasis in pregnancy. Clin Endocrinol (Oxf). 1996; 45, 16.Google Scholar
19. Kovacs, CS, Lanske, B, Hunzelman, JL, et al. Parathyroid hormone-related peptide (PTHrP) regulates fetal-placental calcium transport through a receptor distinct from the PTH/PTHrP receptor. Proc Natl Acad Sci USA. 1996; 93, 1523315238.Google Scholar
20. Namgung, R, Tsang, RC, Li, C, et al. Low total body bone mineral content and high bone resorption in Korean winter-born versus summer-born newborn infants. J Paed. 1998; 132, 285288.Google Scholar
21. Specker, BL, Namgung, R, Tsang, RC. Bone mineral acquisition in utero, during infancy, and throughout childhood. In Osteoporosis (eds. Marcus R, Feldman D, Kelsey J), 2nd edn, 2001; pp. 599620. Academic Press, New York.Google Scholar
22. Cooper, C, Cawley, MID, Bhalla, A, et al. Childhood growth, physical activity and peak bone mass in women. J Bone Min Res. 1995; 10, 940947.Google Scholar
23. Cooper, C, Fall, C, Egger, P, et al. Growth in infancy and bone mass in later life. Ann Rheum Dis. 1997; 56, 1721.Google Scholar
24. Dennison, EM, Syddall, HE, Aihie Sayer, A, Gilbody, HJ, Cooper, C. Birthweight and weight at one year are independent determinants of bone mass in the seventh decade: the Hertfordshire cohort study. Ped Res. 2005; 57, 582586.Google Scholar
25. Yarbrough, DE, Barrett-Connor, E, Morton, DJ. Birth weight as a predictor of adult bone mass in postmenopausal women: the Rancho Bernardo Study. Osteoporosis Int. 2000; 11, 626630.Google Scholar
26. Jones, G, Riley, M, Dwyer, T. Maternal smoking during pregnancy, growth and bone mass in prepubertal children. J Bone Miner Res. 1999; 14, 146151.Google Scholar
27. Javaid, MK, Lekamwasam, S, Clark, J, et al. Infant growth influences proximal femoral geometry in adulthood. J Bone Miner Res. 2006; 21, 508512.Google Scholar
28. Oliver, H, Jameson, K, Aihie Sayer, A, Cooper, C, Dennison, EM. Growth in early life predict bone strength in late adulthood: the Hertfordshire cohort study. Bone. 2007; 41, 400405.Google Scholar
29. Dennison, EM, Arden, NK, Keen, RW, et al. Birthweight, vitamin D receptor genotype and the programming of osteoporosis. Paed Peri Epidemiol. 2001; 15, 211219.Google Scholar
30. Dennison, E, Syddall, H, Rodriguez, S, et al. Polymorphism in the growth hormone gene, weight in infancy and adult bone mass. J Clin Endocrinol Metab. 2004; 89, 48984903.Google Scholar
31. Fall, C, Hindmarsh, P, Dennison, E, et al. Programming of growth hormone secretion and bone mineral density in elderly men; an hypothesis. J Clin Endocrinol Metab. 1998; 83, 135139.Google Scholar
32. Dennison, E, Hindmarsh, P, Fall, C, et al. Profiles of endogenous circulating cortisol and bone mineral density in healthy elderly men. J Clin Endocrinol Metab. 1999; 84, 30583063.Google Scholar
33. Dennison, E, Godfrey, K, Wheeler, T, et al. Determinants of neonatal bone mass. Bone. 1997; 20, 26S.Google Scholar
34. Godfrey, K, Walker-Bone, K, Robinson, S, et al. Neonatal bone mass: influence of parental birthweight, maternal smoking, body composition, and activity during pregnancy. J Bone Min Res. 2001; 16, 16941703.Google Scholar
35. Harvey, NC, Javaid, MK, Poole, JR, et al. Paternal skeletal size predicts intrauterine bone mineral accrual. J Clin Endocrinol Metab. 2008; 93, 16761681.Google Scholar
36. Cole, Z, Gale, C, Javaid, MK, et al. Maternal dietary patterns during pregnancy and childhood bone mass: a longitudinal study. J Bone Miner Res. 2009; 24, 663668.Google Scholar
37. Javaid, MK, Crozier, SR, Harvey, NC, et al. Maternal vitamin D status during pregnancy and childhood bone mass at age 9 years: a longitudinal study. Lancet. 2006; 367, 3643.Google Scholar
38. Javaid, MK, Crozier, SR, Harvey, NC, et al. Maternal and seasonal predictors of change in calcaneal quantitative ultrasound during pregnancy. J Clin Endocrinol Metab. 2005; 90, 51825187.Google Scholar
39. Martin, R, Harvey, NC, Crozier, SR, et al. Placental calcium transporter (PMCA3) gene expression predicts intrauterine bone mineral accrual. Bone. 2007; 40, 12031208.Google Scholar
40. Cooper, C, Eriksson, JG, Forsén, T, et al. Maternal height, childhood growth and risk of hip fracture in later life: a longitudinal study. Osteoporosis Int. 2001; 12, 623629.Google Scholar
41. Ammann, P, Bourrin, S, Bonjour, JP, Meyer, JM, Rizzoli, R. Protein undernutrition-induced bone loss is associated with decreased IGF-1 levels and estrogen deficiency. J Bone Miner Res. 2000; 15, 683690.Google Scholar
42. Mehta, G, Roach, HI, Langley-Evans, S, et al. Intrauterine exposure to a maternal low protein diet reduces adult bone mass and alters growth plate morphology in rats. Calcif Tissue Int. 2002; 71, 493498.Google Scholar
43. Brown, SA, Rogers, LK, Dunn, JK, Gotto, AM, Patsch, W. Development of cholesterol homeostatic memory in the rat is influenced by maternal diets. Metabolism. 1990; 39, 468473.Google Scholar
44. McLeod, KI, Goldrick, RB, Whyte, HM. The effect of maternal malnutrition on the progeny in the rat: studies on growth, body composition and organ cellularity in first and second generation progeny. Aust J Exp Biol Med Sci. 1972; 50, 435446.Google Scholar
45. Snoeck, A, Remacle, C, Reusens, B, Hoet, JJ. Effect of a low protein diet during pregnancy on the fetal rat endocrine pancreas. Biol Neonate. 1990; 57, 107118.Google Scholar
46. Godfrey, KM, Barker, DJP, Osmond, C. Disproportionate fetal growth and raised IGE concentration in adult life. Clin Exp Allergy. 1994; 24, 641648.Google Scholar
47. Weiler, HA, Yuen, CK, Seshia, MM. Growth and bone mineralization of young adults weighing less than 1500 g at birth. Early Human Development. 2002; 67, 101112.Google Scholar
48. Fewtrell, MS, Prentice, A, Jones, SC, Bishop, NJ, Stirling, D, et al. Bone mineralization and turnover in preterm infants at 8–12 years of age: the effect of early diet. J Bone Miner Res. 1999; 14, 810820.Google Scholar
49. Wang, D, Vandermeulen, J, Atkinson, SA. Early life factors predict abnormal growth and bone accretion at prepuberty in former premature infants with/without neonatal dexamethasone exposure. Pediatr Res. 2007; 61, 111116.Google Scholar
50. Gale, CR, Martyn, CN, Kellingray, S, Eastell, R, Cooper, C. Intrauterine programming of adult body composition. J Clin Endocrinol Metab. 2001; 86, 267272.Google Scholar