3,756
Views
36
CrossRef citations to date
0
Altmetric
Review Article

Genetic, metabolic and endocrine aspect of intrauterine growth restriction: an update

, &
Pages 2263-2275 | Received 09 Aug 2016, Accepted 03 Oct 2016, Published online: 26 Oct 2016

References

  • Battaglia FC, Lubchenco LO. A practical classification of newborn infants by weight and gestational age. J Pediatr 1967;71:159–63
  • Small-for-Gestational-Age Fetus, Investigation and Management (Green-top Guideline No. 31) [Internet]. Royal College of Obstetricians & Gynecologists. Available from: https://www.rcog.org.uk/en/guidelines-research-services/guidelines/gtg31/ [last accessed 17 Dec 2015]
  • Figueras F, Gardosi J. Intrauterine growth restriction: new concepts in antenatal surveillance, diagnosis, and management. Am J Obstet Gynecol 2011;204:288–300
  • Figueras F, Gratacos E. Stage-based approach to the management of fetal growth restriction. Prenat Diagn 2014;34:655–9
  • Barker DJ, Osmond C, Golding J, et al. Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ 1989;298:564–7
  • Barker DJP. The origins of the developmental origins theory. J Intern Med 2007;261:412–17
  • Sharma D, Shastri S, Farahbakhsh N, Sharma P. Intrauterine growth restriction – Part 1. J Matern-Fetal Neonatal Med 2016;29:3977–87
  • Sharma D, Farahbakhsh N, Shastri S, Sharma P. Intrauterine growth restriction - Part 2. J Matern-Fetal Neonatal Med 2016;29:4037–48
  • Sharma D, Shastri S, Sharma P. Intrauterine growth restriction: antenatal and postnatal aspects. Clin Med Insights Pediatr 2016;10:67–83
  • Murki S, Sharma D. Intrauterine growth retardation – a review article. J Neonatal Biol. 2014;3:135. doi: 10.4172/2167-0897.1000135
  • Dessì A, Ottonello G, Fanos V. Physiopathology of intrauterine growth retardation: from classic data to metabolomics. J Matern-Fetal Neonatal Med 2012;25:13–18
  • Prince V. The Hox Paradox: more complex(es) than imagined. Dev Biol 2002;249:1–15
  • Hemberger M, Cross JC. Genes governing placental development. Trends Endocrinol Metab TEM 2001;12:162–8
  • Cross JC, Anson-Cartwright L, Scott IC. Transcription factors underlying the development and endocrine functions of the placenta. Recent Prog Horm Res 2002;57:221–34
  • Murthi P. Review: placental homeobox genes and their role in regulating human fetal growth. Placenta 2014;35:S46–50
  • Murthi P, Doherty V, Said J, et al. Homeobox gene HLX1 expression is decreased in idiopathic human fetal growth restriction. Am J Pathol 2006;168:511–18
  • Murthi P, Abumaree M, Kalionis B. Analysis of homeobox gene action may reveal novel angiogenic pathways in normal placental vasculature and in clinical pregnancy disorders associated with abnormal placental angiogenesis. Front Pharmacol 2014;5:133
  • Murthi P, Rajaraman G, Brennecke SP, Kalionis B. The role of placental homeobox genes in human fetal growth restriction. J Pregnancy 2011;2011:548171
  • Murthi P, Doherty VL, Said JM, et al. Homeobox gene ESX1L expression is decreased in human pre-term idiopathic fetal growth restriction. Mol Hum Reprod 2006;12:335–40
  • Silverman GA, Bird PI, Carrell RW, et al. The serpins are an expanding superfamily of structurally similar but functionally diverse proteins. Evolution, mechanism of inhibition, novel functions, and a revised nomenclature. J Biol Chem 2001;276:33293–6
  • Chelbi ST, Mondon F, Jammes H, et al. Expressional and epigenetic alterations of placental serine protease inhibitors: SERPINA3 is a potential marker of preeclampsia. Hypertens (Dallas, TX 1979) 2007;49:76–83
  • Chelbi ST, Wilson ML, Veillard A-C, et al. Genetic and epigenetic mechanisms collaborate to control SERPINA3 expression and its association with placental diseases. Hum Mol Genet 2012;21:1968–78
  • Dealy MJ, Nguyen KV, Lo J, et al. Loss of Cul1 results in early embryonic lethality and dysregulation of cyclin E. Nat Genet 1999;23:245–8
  • Li B, Ruiz JC, Chun KT. CUL-4A is critical for early embryonic development. Mol Cell Biol 2002;22:4997–5005
  • Gascoin-Lachambre G, Buffat C, Rebourcet R, et al. Cullins in human intra-uterine growth restriction: expressional and epigenetic alterations. Placenta 2010;31:151–7
  • van Dijk M, Mulders J, Poutsma A, et al. Maternal segregation of the Dutch preeclampsia locus at 10q22 with a new member of the winged helix gene family. Nat Genet 2005;37:514–19
  • van Dijk M, Oudejans CB. STOX1: key player in trophoblast dysfunction underlying early onset preeclampsia with growth retardation. J Pregnancy. 2011;2011:521826. doi: 10.1155/2011/521826
  • Berends AL, Bertoli-Avella AM, de Groot CJM, et al. STOX1 gene in pre-eclampsia and intrauterine growth restriction. BJOG Int J Obstet Gynaecol 2007;114:1163–7
  • Bond CS, Fox AH. Paraspeckles: nuclear bodies built on long noncoding RNA. J Cell Biol 2009;186:637–44
  • Clemson CM, Hutchinson JN, Sara SA, et al. An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol Cell 2009;33:717–26
  • Gremlich S, Damnon F, Reymondin D, et al. The long non-coding RNA NEAT1 is increased in IUGR placentas, leading to potential new hypotheses of IUGR origin/development. Placenta 2014;35:44–9
  • Iyer S, Leonidas DD, Swaminathan GJ, et al. The crystal structure of human placenta growth factor-1 (PlGF-1), an angiogenic protein, at 2.0 A resolution. J Biol Chem 2001;276:12153–61
  • Maglione D, Guerriero V, Viglietto G, et al. Two alternative mRNAs coding for the angiogenic factor, placenta growth factor (PlGF), are transcribed from a single gene of chromosome 14. Oncogene 1993;8:925–31
  • Li S-W, Ling Y, Jin S, et al. Expression of soluble vascular endothelial growth factor receptor-1 and placental growth factor in fetal growth restriction cases and intervention effect of tetramethylpyrazine. Asian Pac J Trop Med 2014;7:663–7
  • Vrachnis N, Kalampokas E, Sifakis S, et al. Placental growth factor (PlGF): a key to optimizing fetal growth. J Matern-Fetal Neonatal Med 2013;26:995–1002
  • Thadhani R, Mutter WP, Wolf M, et al. First trimester placental growth factor and soluble fms-like tyrosine kinase 1 and risk for preeclampsia. J Clin Endocrinol Metab 2004;89:770–5
  • Taylor RN, Grimwood J, Taylor RS, et al. Longitudinal serum concentrations of placental growth factor: evidence for abnormal placental angiogenesis in pathologic pregnancies. Am J Obstet Gynecol 2003;188:177–82
  • Stewart PM, Krozowski ZS. 11beta-Hydroxysteroid dehydrogenase. Vitam Horm 1999;57:249–324
  • Sun K, Yang K, Challis JR. Differential expression of 11 beta-hydroxysteroid dehydrogenase types 1 and 2 in human placenta and fetal membranes. J Clin Endocrinol Metab 1997;82:300–5
  • Wächter R, Masarik L, Bürzle M, et al. Differential expression and activity of 11beta-hydroxysteroid dehydrogenase in human placenta and fetal membranes from pregnancies with intrauterine growth restriction. Fetal Diagn Ther 2009;25:328–35
  • Tzschoppe A, Struwe E, Blessing H, et al. Placental 11beta-HSD2 gene expression at birth is inversely correlated with growth velocity in the first year of life after intrauterine growth restriction. Pediatr Res 2009;65:647–53
  • Ward RM. Pharmacologic enhancement of fetal lung maturation. Clin Perinatol 1994;21:523–42
  • Börzsönyi B, Demendi C, Pajor A, et al. Gene expression patterns of the 11β-hydroxysteroid dehydrogenase 2 enzyme in human placenta from intrauterine growth restriction: the role of impaired feto-maternal glucocorticoid metabolism. Eur J Obstet Gynecol Reprod Biol 2012;161:12–17
  • Shams M, Kilby MD, Somerset DA, et al. 11Beta-hydroxysteroid dehydrogenase type 2 in human pregnancy and reduced expression in intrauterine growth restriction. Hum Reprod Oxf Engl 1998;13:799–804
  • Daher S, Guimarães AJ, Mattar R, et al. Bcl-2 and Bax expressions in pre-term, term and post-term placentas. Am J Reprod Immunol N Y N 1989 2008;60:172–8
  • Endo H, Okamoto A, Yamada K, et al. Frequent apoptosis in placental villi from pregnancies complicated with intrauterine growth restriction and without maternal symptoms. Int J Mol Med 2005;16:79–84
  • Börzsönyi B, Demendi C, Rigó J, et al. The regulation of apoptosis in intrauterine growth restriction: a study of Bcl-2 and Bax gene expression in human placenta. J Matern-Fetal Neonatal Med 2013;26:347–50
  • Jones JI, Clemmons DR. Insulin-like growth factors and their binding proteins: biological actions. Endocr Rev 1995;16:3–34
  • Wang HS, Chard T. The role of insulin-like growth factor-I and insulin-like growth factor-binding protein-1 in the control of human fetal growth. J Endocrinol 1992;132:11–19
  • Ashton IK, Zapf J, Einschenk I, MacKenzie IZ. Insulin-like growth factors (IGF) 1 and 2 in human foetal plasma and relationship to gestational age and foetal size during midpregnancy. Acta Endocrinol (Copenh) 1985;110:558–63
  • Börzsönyi B, Demendi C, Nagy Z, et al. Gene expression patterns of insulin-like growth factor 1, insulin-like growth factor 2 and insulin-like growth factor binding protein 3 in human placenta from pregnancies with intrauterine growth restriction. J Perinat Med 2011;39:701–7
  • Lee MH, Jeon YJ, Lee SM, et al. Placental gene expression is related to glucose metabolism and fetal cord blood levels of insulin and insulin-like growth factors in intrauterine growth restriction. Early Hum Dev 2010;86:45–50
  • Scott J, Urdea M, Quiroga M, et al. Structure of a mouse submaxillary messenger RNA encoding epidermal growth factor and seven related proteins. Science 1983;221:236–40
  • Morrish DW, Bhardwaj D, Dabbagh LK, et al. Epidermal growth factor induces differentiation and secretion of human chorionic gonadotropin and placental lactogen in normal human placenta. J Clin Endocrinol Metab 1987;65:1282–90
  • Casalini P, Iorio MV, Galmozzi E, Ménard S. Role of HER receptors family in development and differentiation. J Cell Physiol 2004;200:343–50
  • Rab A, Szentpéteri I, Kornya L, et al. Placental gene expression patterns of epidermal growth factor in intrauterine growth restriction. Eur J Obstet Gynecol Reprod Biol 2013;170:96–9
  • Robinson CJ, Johnson DD. Soluble endoglin as a second-trimester marker for preeclampsia. Am J Obstet Gynecol 2007;197:174.e1–5
  • Smith GCS, Wear H. The perinatal implications of angiogenic factors. Curr Opin Obstet Gynecol 2009;21:111–16
  • ten Dijke P, Goumans M-J, Pardali E. Endoglin in angiogenesis and vascular diseases. Angiogenesis 2008;11:79–89
  • Wipff J, Avouac J, Borderie D, et al. Disturbed angiogenesis in systemic sclerosis: high levels of soluble endoglin. Rheumatology (Oxford) 2008;47:972–5
  • Fujishita A, Hasuo A, Khan KN, et al. Immunohistochemical study of angiogenic factors in endometrium and endometriosis. Gynecol Obstet Invest 1999;48:36–44
  • Baumwell S, Karumanchi SA. Pre-eclampsia: clinical manifestations and molecular mechanisms. Nephron Clin Pract 2007;106:c72–81
  • Venkatesha S, Toporsian M, Lam C, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med 2006;12:642–9
  • Szentpéteri I, Rab A, Kornya L, et al. Placental gene expression patterns of endoglin (CD105) in intrauterine growth restriction. J Matern-Fetal Neonatal Med 2014;27:350–4
  • Jeyabalan A, McGonigal S, Gilmour C, et al. Circulating and placental endoglin concentrations in pregnancies complicated by intrauterine growth restriction and preeclampsia. Placenta 2008;29:555–63
  • Yinon Y, Nevo O, Xu J, et al. Severe intrauterine growth restriction pregnancies have increased placental endoglin levels: hypoxic regulation via transforming growth factor-beta 3. Am J Pathol 2008;172:77–85
  • Mayhew TM, Charnock-Jones DS, Kaufmann P. Aspects of human fetoplacental vasculogenesis and angiogenesis. III. Changes in complicated pregnancies. Placenta 2004;25:127–39
  • Chen C-P, Bajoria R, Aplin JD. Decreased vascularization and cell proliferation in placentas of intrauterine growth-restricted fetuses with abnormal umbilical artery flow velocity waveforms. Am J Obstet Gynecol 2002;187:764–9
  • Gourvas V, Dalpa E, Konstantinidou A, et al. Angiogenic factors in placentas from pregnancies complicated by fetal growth restriction (review). Mol Med Rep 2012;6:23–7
  • Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev 2004;25:581–611
  • Ferrara N. Role of vascular endothelial growth factor in regulation of physiological angiogenesis. Am J Physiol Cell Physiol 2001;280:C1358–66
  • Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669–76
  • Hood JD, Meininger CJ, Ziche M, Granger HJ. VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells. Am J Physiol 1998;274:H1054–8
  • Szentpéteri I, Rab A, Kornya L, et al. Gene expression patterns of vascular endothelial growth factor (VEGF-A) in human placenta from pregnancies with intrauterine growth restriction. J Matern-Fetal Neonatal Med 2013;26:984–9
  • Lash G, MacPherson A, Liu D, et al. Abnormal fetal growth is not associated with altered chorionic villous expression of vascular endothelial growth factor mRNA. Mol Hum Reprod 2001;7:1093–8
  • Lyall F, Young A, Boswell F, et al. Placental expression of vascular endothelial growth factor in placentae from pregnancies complicated by pre-eclampsia and intrauterine growth restriction does not support placental hypoxia at delivery. Placenta 1997;18:269–76
  • Caniggia I, Winter JL. Adriana and Luisa Castellucci Award lecture 2001. Hypoxia inducible factor-1: oxygen regulation of trophoblast differentiation in normal and pre-eclamptic pregnancies-a review. Placenta 2002;23:S47–57
  • Graham CH, Lysiak JJ, McCrae KR, Lala PK. Localization of transforming growth factor-beta at the human fetal–maternal interface: role in trophoblast growth and differentiation. Biol Reprod 1992;46:561–72
  • Chen H-H, Zhao S, Song J-G. TGF-beta1 suppresses apoptosis via differential regulation of MAP kinases and ceramide production. Cell Death Differ 2003;10:516–27
  • Chauvin S, Yinon Y, Xu J, et al. Aberrant TGFβ signalling contributes to dysregulation of sphingolipid metabolism in intrauterine growth restriction. J Clin Endocrinol Metab 2015;100:E986–96
  • Leduc L, Levy E, Bouity-Voubou M, Delvin E. Fetal programming of atherosclerosis: possible role of the mitochondria. Eur J Obstet Gynecol Reprod Biol 2010;149:127–30
  • Lattuada D, Colleoni F, Martinelli A, et al. Higher mitochondrial DNA content in human IUGR placenta. Placenta 2008;29:1029–33
  • Colleoni F, Lattuada D, Garretto A, et al. Maternal blood mitochondrial DNA content during normal and intrauterine growth restricted (IUGR) pregnancy. Am J Obstet Gynecol 2010;203:365.e1–6
  • Mando’ C, Marino MA, Miriam F, et al. OS048 mitochondrial content and function in placental cells and tissues of preeclampsia and IUGR. Pregnancy Hypertens 2012;2:203
  • Mandò C, Palma CD, Stampalija T, et al. Placental mitochondrial content and function in intrauterine growth restriction and preeclampsia. Am J Physiol – Endocrinol Metab 2014;306:E404–13
  • Chan JCN, Cheung JCK, Stehouwer CDA, et al. The central roles of obesity-associated dyslipidaemia, endothelial activation and cytokines in the metabolic syndrome – an analysis by structural equation modeling. Int J Obes Relat Metab Disord J Int Assoc Study Obes 2002;26:994–1008
  • Kupferminc MJ, Peaceman AM, Wigton TR, et al. Tumor necrosis factor-alpha is elevated in plasma and amniotic fluid of patients with severe preeclampsia. Am J Obstet Gynecol 1994;170:1752–1759
  • Haeger M, Unander M, Andersson B, et al. Increased release of tumor necrosis factor-alpha and interleukin-6 in women with the syndrome of hemolysis, elevated liver enzymes, and low platelet count. Acta Obstet Gynecol Scand 1996;75:695–701
  • Molvarec A, Jermendy A, Nagy B, et al. Association between tumor necrosis factor (TNF)-alpha G-308A gene polymorphism and preeclampsia complicated by severe fetal growth restriction. Clin Chim Acta Int J Clin Chem 2008;392:52–7
  • Folz RJ, Crapo JD. Extracellular superoxide dismutase (SOD3): tissue-specific expression, genomic characterization, and computer-assisted sequence analysis of the human EC SOD gene. Genomics 1994;22:162–71
  • Rumiris D, Purwosunu Y, Wibowo N, et al. Lower rate of preeclampsia after antioxidant supplementation in pregnant women with low antioxidant status. Hypertens Pregnancy 2006;25:241–53
  • Rosta K, Molvarec A, Enzsöly A, et al. Association of extracellular superoxide dismutase (SOD3) Ala40Thr gene polymorphism with pre-eclampsia complicated by severe fetal growth restriction. Eur J Obstet Gynecol Reprod Biol 2009;142:134–8
  • Yanagisawa M, Kurihara H, Kimura S, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature 1988;332:411–15
  • Di Iorio R, Marinoni E, Anceschi MM, et al. Amniotic fluid endothelin-1 levels are increased in pregnancy-induced hypertension and intrauterine growth retardation. Am J Reprod Immunol (N Y N 1989) 1996;36:260–3
  • Liu YA, Ostlund E, Fried G. Endothelin-induced contractions in human placental blood vessels are enhanced in intrauterine growth retardation, and modulated by agents that regulate levels of intracellular calcium. Acta Physiol Scand 1995;155:405–14
  • Pelleymounter MA, Cullen MJ, Baker MB, et al. Effects of the obese gene product on body weight regulation in ob/ob mice. Science 1995;269:540–3
  • Remesar X, Rafecas I, Fernández-López JA, Alemany M. Leptin. Med Res Rev 1997;17:225–34
  • Arslan M, Yazici G, Erdem A, et al. Endothelin 1 and leptin in the pathophysiology of intrauterine growth restriction. Int J Gynaecol Obstet 2004;84:120–6
  • Nezar MA-S, el-Baky AMA, Soliman OA-S, et al. Endothelin-1 and leptin as markers of intrauterine growth restriction. Indian J Pediatr 2009;76:485–8
  • Wang C-N, Chang S-D, Peng H-H, et al. Change in amniotic fluid levels of multiple anti-angiogenic proteins before development of preeclampsia and intrauterine growth restriction. J Clin Endocrinol Metab 2010;95:1431–41
  • Skoczylas A. The role of visfatin in the pathophysiology of human. Wiad Lek Wars (Pol 1960) 2009;62:190–6
  • Malamitsi-Puchner A, Briana DD, Boutsikou M, et al. Perinatal circulating visfatin levels in intrauterine growth restriction. Pediatrics 2007;119:e1314–18
  • Laskowska M, Laskowska K, Oleszczuk J. Endoglin in pregnancy complicated by fetal intrauterine growth restriction in normotensive and preeclamptic pregnant women: a comparison between preeclamptic patients with appropriate-for-gestational-age weight infants and healthy pregnant women. J Matern-Fetal Neonatal Med 2012;25:806–11
  • Gearing AJ, Newman W. Circulating adhesion molecules in disease. Immunol Today 1993;14:506–12
  • Springer TA. Adhesion receptors of the immune system. Nature 1990;346:425–34
  • Coata G, Pennacchi L, Bini V, et al. Soluble adhesion molecules: marker of pre-eclampsia and intrauterine growth restriction. J Matern Fetal Neonatal Med 2002;12:28–34
  • Djurovic S, Schjetlein R, Wisløff F, et al. Increased levels of intercellular adhesion molecules and vascular cell adhesion molecules in pre-eclampsia. Br J Obstet Gynaecol 1997;104:466–70
  • Bretelle F, Sabatier F, Blann A, et al. Maternal endothelial soluble cell adhesion molecules with isolated small for gestational age fetuses: comparison with pre-eclampsia. BJOG Int J Obstet Gynaecol 2001;108:1277–82
  • Kupferminc MJ, Eldor A, Steinman N, et al. Increased frequency of genetic thrombophilia in women with complications of pregnancy. N Engl J Med 1999;340:9–13
  • Bertina RM, Koeleman BP, Koster T, et al. Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature 1994;369:64–7
  • Grandone E, Margaglione M, Colaizzo D, et al. Lower birth-weight in neonates of mothers carrying factor V G1691A and factor II A(20210) mutations. Haematologica 2002;87:177–81
  • von Kries R, Junker R, Oberle D, et al. Foetal growth restriction in children with prothrombotic risk factors. Thromb Haemost 2001;86:1012–6
  • Oh-I S, Shimizu H, Satoh T, et al. Identification of nesfatin-1 as a satiety molecule in the hypothalamus. Nature 2006;443:709–12
  • Aydin S. Multi-functional peptide hormone NUCB2/nesfatin-1. Endocrine 2013;44:312–25
  • Pałasz A, Krzystanek M, Worthington J, et al. Nesfatin-1, a unique regulatory neuropeptide of the brain. Neuropeptides 2012;46:105–12
  • Serin S, Bakacak M, Ercan Ö, et al. The evaluation of Nesfatin-1 levels in patients with and without intrauterine growth restriction. J Matern Fetal Neonatal Med 2016;29:1409–13
  • Buhimschi IA, Saade GR, Chwalisz K, Garfield RE. The nitric oxide pathway in pre-eclampsia: pathophysiological implications. Hum Reprod Update 1998;4:25–42
  • Andersen GN, Caidahl K, Kazzam E, et al. Correlation between increased nitric oxide production and markers of endothelial activation in systemic sclerosis: findings with the soluble adhesion molecules E-selectin, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1. Arthritis Rheum 2000;43:1085–93
  • Holden DP, Fickling SA, Whitley GS, Nussey SS. Plasma concentrations of asymmetric dimethylarginine, a natural inhibitor of nitric oxide synthase, in normal pregnancy and preeclampsia. Am J Obstet Gynecol 1998;178:551–6
  • Savvidou MD, Hingorani AD, Tsikas D, et al. Endothelial dysfunction and raised plasma concentrations of asymmetric dimethylarginine in pregnant women who subsequently develop pre-eclampsia. Lancet Lond Engl 2003;361:1511–17
  • Laskowska M, Laskowska K, Leszczyńska-Gorzelak B, Oleszczuk J. Asymmetric dimethylarginine in normotensive pregnant women with isolated fetal intrauterine growth restriction: a comparison with preeclamptic women with and without intrauterine growth restriction. J Matern Fetal Neonatal Med 2011;24:936–42
  • Miozzo M, Simoni G. The role of imprinted genes in fetal growth. Biol Neonate 2002;81:217–28
  • Hannula K, Lipsanen-Nyman M, Kontiokari T, Kere J. A narrow segment of maternal uniparental disomy of chromosome 7q31-qter in Silver-Russell syndrome delimits a candidate gene region. Am J Hum Genet 2001;68:247–53
  • Blagitko N, Mergenthaler S, Schulz U, et al. Human GRB10 is imprinted and expressed from the paternal and maternal allele in a highly tissue-and isoform-specific fashion. Hum Mol Genet 2000;9:1587–95
  • Langlois S, Yong SL, Wilson RD, et al. Prenatal and postnatal growth failure associated with maternal heterodisomy for chromosome 7. J Med Genet 1995;32:871–5
  • Miozzo M, Grati FR, Bulfamante G, et al. Post-zygotic origin of complete maternal chromosome 7 isodisomy and consequent loss of placental PEG1/MEST expression. Placenta 2001;22:813–21
  • Heizmann CW. Ca2+-binding S100 proteins in the central nervous system. Neurochem Res 1999;24:1097–100
  • Gazzolo D, Marinoni E, di Iorio R, et al. Circulating S100beta protein is increased in intrauterine growth-retarded fetuses. Pediatr Res 2002;51:215–19
  • Florio P, Marinoni E, Di Iorio R, et al. Urinary S100B protein concentrations are increased in intrauterine growth-retarded newborns. Pediatrics 2006;118:e747–54
  • Philbrick WM, Wysolmerski JJ, Galbraith S, et al. Defining the roles of parathyroid hormone-related protein in normal physiology. Physiol Rev 1996;76:127–73
  • Briana DD, Boutsikou M, Baka S, et al. N-terminal parathyroid hormone-related protein levels in human intrauterine growth restricted pregnancies. Acta Obstet Gynecol Scand 2007;86:945–9
  • Curtis NE, King RG, Moseley JM, et al. Preterm fetal growth restriction is associated with increased parathyroid hormone-related protein expression in the fetal membranes. Am J Obstet Gynecol 2000;183:700–5
  • Ichiki Y, Kitamura K, Kangawa K, et al. Distribution and characterization of immunoreactive adrenomedullin in human tissue and plasma. FEBS Lett 1994;338:6–10
  • Di Iorio R, Marinoni E, Letizia C, et al. Adrenomedullin is increased in the fetoplacental circulation in intrauterine growth restriction with abnormal umbilical artery waveforms. Am J Obstet Gynecol 2000;182:650–4
  • Marchini A, Ogata T, Rappold GA. A track record on SHOX: from basic research to complex models and therapy. Endocr Rev 2016;37:417–48
  • Caliebe J, Broekman S, Boogaard M, et al. IGF1, IGF1R and SHOX mutation analysis in short children born small for gestational age and short children with normal birth size (idiopathic short stature). Horm Res Pædiatrics 2012;77:250–60
  • LeRoith D, Werner H, Beitner-Johnson D, Roberts CT. Molecular and cellular aspects of the insulin-like growth factor I receptor. Endocr Rev 1995;16:143–63
  • Kawashima Y, Higaki K, Fukushima T, et al. Novel missense mutation in the IGF-I receptor L2 domain results in intrauterine and postnatal growth retardation. Clin Endocrinol (Oxf) 2012;77:246–54
  • Leal AC, Montenegro LR, Saito RF, et al. Analysis of the insulin-like growth factor 1 receptor gene in children born small for gestational age: in vitro characterization of a novel mutation (p.Arg511Trp). Clin Endocrinol (Oxf) 2013;78:558–63
  • Leal ADC, Canton APM, Montenegro LR, et al. [Mutations in insulin-like growth factor receptor 1 gene (IGF1R) resulting in intrauterine and postnatal growth retardation]. Arq Bras Endocrinol Metabol 2011;55:541–9
  • Abuzzahab MJ, Schneider A, Goddard A, et al. IGF-I receptor mutations resulting in intrauterine and postnatal growth retardation. N Engl J Med 2003;349:2211–22
  • Akturk A, Onal EE, Atalay Y, et al. Maternal and umbilical venous adrenomedullin and nitric oxide levels in intrauterine growth restriction. J Matern-Fetal Neonatal Med 2007;20:521–5
  • Tranquilli AL, Bezzeccheri V, Giannubilo SR, et al. Amniotic levels of nitric oxide in women with fetal intrauterine growth restriction. J Matern Fetal Neonatal Med 2003;13:115–18
  • Devaskar SU, Raychaudhuri S. Epigenetics-a science of heritable biological adaptation. Pediatr Res 2007;61:1R–4R
  • Laird PW. Cancer epigenetics. Hum Mol Genet 2005;14:R65–76
  • Dolinoy DC, Das R, Weidman JR, Jirtle RL. Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr Res 2007;61:30R–7R
  • Nawathe AR, Christian M, Kim SH, et al. Insulin-like growth factor axis in pregnancies affected by fetal growth disorders. Clin Epigenetics 2016;8:11
  • Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell 2009;136:642–55
  • Mouillet J-F, Chu T, Hubel CA, et al. The levels of hypoxia-regulated microRNAs in plasma of pregnant women with fetal growth restriction. Placenta 2010;31:781–4
  • Tang Q, Wu W, Xu X, et al. miR-141 contributes to fetal growth restriction by regulating PLAG1 expression. PLoS One 2013;8:e58737
  • Huang L, Shen Z, Xu Q, et al. Increased levels of microRNA-424 are associated with the pathogenesis of fetal growth restriction. Placenta 2013;34:624–7
  • Higashijima A, Miura K, Mishima H, et al. Characterization of placenta-specific microRNAs in fetal growth restriction pregnancy. Prenat Diagn 2013;33:214–22
  • Cetin I, Alvino G. Intrauterine growth restriction: implications for placental metabolism and transport. A review. Placenta 2009;30:S77–82
  • Jansson T, Powell TL. IFPA 2005 Award in Placentology Lecture. Human placental transport in altered fetal growth: does the placenta function as a nutrient sensor? – a review. Placenta 2006;27:S91–7
  • Sibley CP, Turner MA, Cetin I, et al. Placental phenotypes of intrauterine growth. Pediatr Res 2005;58:827–32
  • Marconi AM, Cetin I, Davoli E, et al. An evaluation of fetal glucogenesis in intrauterine growth-retarded pregnancies. Metab Clin Exp 1993;42:860–4
  • Marconi AM, Paolini C, Buscaglia M, et al. The impact of gestational age and fetal growth on the maternal–fetal glucose concentration difference. Obstet Gynecol 1996;87:937–42
  • Cetin I, Giovannini N, Alvino G, et al. Intrauterine growth restriction is associated with changes in polyunsaturated fatty acid fetal–maternal relationships. Pediatr Res 2002;52:750–5
  • Wadsack C, Tabano S, Maier A, et al. Intrauterine growth restriction is associated with alterations in placental lipoprotein receptors and maternal lipoprotein composition. Am J Physiol Endocrinol Metab 2007;292:E476–84
  • Zhang S, Regnault TRH, Barker PL, et al. Placental adaptations in growth restriction. Nutrients 2015;7:360–89

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.