Placental biomarkers of fetal-originated diseases | Placenta and Reproductive Medicine

Placental biomarkers of fetal-originated diseases

Authors

  • Pengxia Yu
  • Sijia Chen
  • Hui Wang Department of Pharmacology, Basic Medical School of Wuhan University

DOI:

https://doi.org/10.54844/prm.2024.0466

Keywords:

placenta, adverse environment, fetal growth restriction, fetal-originated diseases, biomarkers

Abstract

Fetal-originated diseases refer to birth defects of offspring and multiple chronic diseases in adulthood caused by abnormal embryonic development. However, due to the vulnerability of the fetus/ neonate and technical limitations, it is very difficult to carry out effective development assessment and early warning of long-term diseases in the early stages of life. It is known that the placenta is the unique link between the mother and the fetus, and its role in the occurrence and progression of fetal-originated diseases cannot be ignored. Studies have found that a variety of adverse environmental factors (such as nanoparticle exposure) cannot pass through the placental barrier, but can lead to fetal dysplasia and multi-organ development programming changes by affecting placental development, and ultimately mediate the occurrence of fetal-originated diseases. Meanwhile, some environmental factors that can pass through the placental barrier can cause placenta-fetal co-exposure, resulting in similar signaling pathways and epigenetic changes. The placenta originates from the fetus and the mother, and its development is accompanied by changes in indicators that can be objectively and quantitatively detected. These factors can be used as a biomarker to assess maternal exposure, and placental function, and to predict the developmental status and long-term disease susceptibility of offspring. To date, researchers have discovered a variety of potential placental biomarkers, and show promising application prospects. This paper reviews the recent research on placenta-related mechanisms leading to fetal-originated diseases and placental biomarkers, to provide the theoretical and experimental basis for early warning, prevention, and treatment of fetal-originated diseases.

References

Uauy R, Kain J, Corvalan C. How can the Developmental Origins of Health and Disease (DOHaD) hypothesis contribute to improving health in developing countries? Am J Clin Nutr 2011;94:1759.

Hedderson MM, Ferrara A, Avalos LA, et al. The Kaiser Permanente Northern California research program on genes, environment, and health (RPGEH) pregnancy cohort:study design, methodology and baseline characteristics. BMC Pregnancy Childbirth 2016;16:381.

Fleming TP, Sun C, Denisenko O, et al. Environmental Exposures around Conception:Developmental Pathways Leading to Lifetime Disease Risk. Int J Environ Res Public Health 2021;18:9380.

Drwal E, Rak A, Gregoraszczuk EL. Review:Polycyclic aromatic hydrocarbons (PAHs)-Action on placental function and health risks in future life of newborns. Toxicology 2019;411:133–142.

Morales-Rubio RA, Alvarado-Cruz I, Manzano-León N, et al. In utero exposure to ultrafine particles promotes placental stress-induced programming of renin-angiotensin system-related elements in the offspring results in altered blood pressure in adult mice. Part Fibre Toxicol 2019;16:7.

Ghosh R, Causey K, Burkart K, Wozniak S, Cohen A, Brauer M. Ambient and household PM2.5 pollution and adverse perinatal outcomes:A meta-regression and analysis of attributable global burden for 204 countries and territories. PLoS Med 2021;18:e1003718.

Arias A, Schander JA, Bariani MV, et al. Dexamethasone-induced intrauterine growth restriction modulates expression of placental vascular growth factors and fetal and placental growth. Mol Hum Reprod 2021;27:gaab006.

Bar-Lev MR, Maayan-Metzger A, Matok I, Heyman Z, Sivan E, Kuint J. Short-term outcomes in low birth weight infants following antenatal exposure to betamethasone versus dexamethasone. Obstet Gynecol 2004;104:484–488.

Xiong YW, Zhu HL, Nan Y, et al. Maternal cadmium exposure during late pregnancy causes fetal growth restriction via inhibiting placental progesterone synthesis. Ecotoxicol Environ Saf 2020;187:109879.

Zhu HL, Shi XT, Xu XF, et al. Environmental cadmium exposure induces fetal growth restriction via triggering PERK-regulated mitophagy in placental trophoblasts. Environ Int 2021;147:106319.

Liu L, Liu F, Kou H, et al. Prenatal nicotine exposure induced a hypothalamic-pituitary-adrenal axis-associated neuroendocrine metabolic programmed alteration in intrauterine growth retardation offspring rats. Toxicol Lett 2012;214:307–313.

Feng JH, Yan YE, Liang G, et al. Maternal and fetal metabonomic alterations in prenatal nicotine exposure-induced rat intrauterine growth retardation. Mol Cell Endocrinol 2014;394(1–2):59–69. DOI:10.1016/j.mce.2014.06.016]

Connor S, Tan KY, Pestell CF, Fitzpatrick JP. The Demographic and Neurocognitive Profile of Clients Diagnosed With Fetal Alcohol Spectrum Disorder in PATCHES Paediatrics Clinics Across Western Australia and the Northern Territory. Alcohol Clin Exp Res 2020;44:1284–1291.

He Z, Zhang J, Chen G, et al. H19/let-7 axis mediates caffeine exposure during pregnancy induced adrenal dysfunction and its multi-generation inheritance. Sci Total Environ 2021;792:148440.

Jafari A, Naghshi S, Shahinfar H, et al. Relationship between maternal caffeine and coffee intake and pregnancy loss:A grading of recommendations assessment, development, and evaluation-assessed, dose-response meta-analysis of observational studies. Front Nutr 2022;9:886224.

Janardhan N, Roy PG, Vyas AK. Impact of maternal substance use on offspring's cardiovascular health. Toxicol Appl Pharmacol 2022;450:116164.

Cestonaro C, Menozzi L, Terranova C. Infants of Mothers with Cocaine Use:Review of Clinical and Medico-Legal Aspects. Children (Basel) 2022;9(1):67. [PMID:35053692 DOI:10.3390/children9010067]

Velez ML, Jordan C, Jansson LM. Reconceptualizing non-pharmacologic approaches to Neonatal Abstinence Syndrome (NAS) and Neonatal Opioid Withdrawal Syndrome (NOWS):A theoretical and evidence-based approach. Part II:The clinical application of nonpharmacologic care for NAS/NOWS. Neurotoxicol Teratol 2021;88:107032.

Albano GD, La Spina C, Pitingaro W, et al. Intrauterine and Neonatal Exposure to Opioids:Toxicological, Clinical, and Medico-Legal Issues. Toxics 2023;11:62.

Hawkins SJ, Crompton LA, Sood A, et al. Nanoparticle-induced neuronal toxicity across placental barriers is mediated by autophagy and dependent on astrocytes. Nat Nanotechnol 2018;13:427–433.

Choi H, Rauh V, Garfinkel R, Tu Y, Perera FP. Prenatal exposure to airborne polycyclic aromatic hydrocarbons and risk of intrauterine growth restriction. Environ Health Perspect 2008;116:658–665.

Perera FP, Rauh V, Tsai WY, et al. Effects of transplacental exposure to environmental pollutants on birth outcomes in a multiethnic population. Environ Health Perspect 2003;111:201–205.

Li R, Wang X, Wang B, et al. Gestational 1-nitropyrene exposure causes fetal growth restriction through disturbing placental vascularity and proliferation. Chemosphere 2018;213:252–258.

Yingya Zhao, Qi Song, Wenzhen Ge, et al. Associations between in utero exposure to polybrominated diphenyl ethers, pathophysiological state of fetal growth and placental DNA methylation changes. Environ Int 2019;133:105255.

Jin Y, Li J, Deng X, et al. Association between fetal growth restriction and maternal exposure to polybrominated diphenyl ethers. Ecotoxicol Environ Saf 2020;198:110623.

Yu Z, Han Y, Shen R, et al. Gestational di-(2-ethylhexyl) phthalate exposure causes fetal intrauterine growth restriction through disturbing placental thyroid hormone receptor signaling. Toxicol Lett 2018;294:1–10.

Guo C, Yang Y, Shi MX, et al. Critical time window of fenvalerate-induced fetal intrauterine growth restriction in mice. Ecotoxicol Environ Saf 2019;172:186–193.

Wang B, Liu JJ, Wang Y, et al. Maternal Fenvalerate Exposure Induces Fetal Intrauterine Growth Restriction Through Disrupting Placental Thyroid Hormone Receptor Signaling. Toxicol Sci 2017;157:377–386.

Sabra S, Malmqvist E, Saborit A, Gratacos E, Gomez Roig MD. Heavy metals exposure levels and their correlation with different clinical forms of fetal growth restriction. PLoS One 2017;12:e0185645.

Camsari C, Folger JK, McGee D, et al. Effects of Periconception Cadmium and Mercury Co-Administration to Mice on Indices of Chronic Diseases in Male Offspring at Maturity. Environ Health Perspect 2017;125:643–650.

Müller JE, Meyer N, Santamaria CG, et al. Bisphenol A exposure during early pregnancy impairs uterine spiral artery remodeling and provokes intrauterine growth restriction in mice. Sci Rep 2018;8:9196.

Nava-Castro KE, Ramírez-Nieto R, Méndez-García LA, et al. Environmental Pollution as a Risk Factor in Testicular Tumour Development:Focus on the Interaction between Bisphenol A and the Associated Immune Response. Int J Environ Res Public Health 2019;16:4113.

Levario-Carrillo M, Amato D, Ostrosky-Wegman P, González-Horta C, Corona Y, Sanin LH. Relation between pesticide exposure and intrauterine growth retardation. Chemosphere 2004;55:1421–1427.

Zhao Y, Chen L, Li LX, et al. Gender-specific relationship between prenatal exposure to phthalates and intrauterine growth restriction. Pediatr Res 2014;76:401–408.

Milnerowicz-Nabzdyk E, Bizon A. How does tobacco smoke influence the morphometry of the fetus and the umbilical cord?-Research on pregnant women with intrauterine growth restriction exposed to tobacco smoke. Reprod Toxicol 2015;58:79–84.

Onoda A, Takeda K, Umezawa M. Pretreatment with N-acetyl cysteine suppresses chronic reactive astrogliosis following maternal nanoparticle exposure during gestational period. Nanotoxicology 2017;11:1012–1025.

Carter RC, Wainwright H, Molteno CD, et al. Alcohol, Methamphetamine, and Marijuana Exposure Have Distinct Effects on the Human Placenta. Alcohol Clin Exp Res 2016;40:753–764.

Singh M, Varukolu S, Chauhan A, et al. Paracetamol exposure and asthma:What does the evidence say? An overview of systematic reviews. Pediatr Pulmonol 2021;56:3189–3199.

Ryan L, Ehrlich S, Finnegan L. Cocaine abuse in pregnancy:effects on the fetus and newborn. Neurotoxicol Teratol 1987;9:295–299.

Xiao D, Huang X, Xu Z, Yang S, Zhang L. Prenatal cocaine exposure differentially causes vascular dysfunction in adult offspring. Hypertension 2009;53:937–943.

Pacheco ALD, de Melo IS, de Souza FMA, et al. Maternal crack cocaine use in rats leads to depressive- and anxiety-like behavior, memory impairment, and increased seizure susceptibility in the offspring. Eur Neuropsychopharmacol 2021;44:34–50.

de Castro A, Jones HE, Johnson RE, Gray TR, Shakleya DM, Huestis MA. Maternal methadone dose, placental methadone concentrations, and neonatal outcomes. Clin Chem 2011;57:449–458.

Hulse GK, Milne E, English DR, Holman CD. The relationship between maternal use of heroin and methadone and infant birth weight. Addiction 1997;92(11):1571–1579.

Fishman B, Daniel S, Koren G, Lunenfeld E, Levy A. Pregnancy outcome following opioid exposure:A cohort study. PLoS One 2019;14:e0219061.

Straub HL, Mou J, Drennan KJ, Pflugeisen BM. Maternal Marijuana Exposure and Birth Weight:An Observational Study Surrounding Recreational Marijuana Legalization. Am J Perinatol 2021;38:65–75.

Smid MC, Metz TD, McMillin GA, et al. Prenatal Nicotine or Cannabis Exposure and Offspring Neurobehavioral Outcomes. Obstet Gynecol 2022;139:21–30.

Paul SE, Hatoum AS, Fine JD, et al. Associations Between Prenatal Cannabis Exposure and Childhood Outcomes:Results From the ABCD Study. JAMA Psychiatry 2021;78:64–76.

Doi M, Nakama N, Sumi T, Usui N, Shimada S. Prenatal methamphetamine exposure causes dysfunction in glucose metabolism and low birthweight. Front Endocrinol (Lausanne) 2022;13:1023984.

Dong N, Zhu J, Han W, et al. Maternal methamphetamine exposure causes cognitive impairment and alteration of neurodevelopment-related genes in adult offspring mice. Neuropharmacology 2018;140:25–34.

Sanjari Moghaddam H, Mobarak Abadi M, Dolatshahi M, et al. Effects of Prenatal Methamphetamine Exposure on the Developing Human Brain:A Systematic Review of Neuroimaging Studies. ACS Chem Neurosci 2021;12:2729–2748.

Lu X, Mao T, Dai Y, et al. Azithromycin exposure during pregnancy disturbs the fetal development and its characteristic of multi-organ toxicity. Life Sci 2023;329:121985. [PMID:37516432 DOI:10.1016/j.lfs.2023.121985]

Flory T Muanda, Odile Sheehy, Anick Bérard. Use of antibiotics during pregnancy and risk of spontaneous abortion. CMAJ 2017;189:E625–E633.

Voormolen DN, de Wit L, van Rijn BB, et al. Neonatal Hypoglycemia Following Diet-Controlled and Insulin-Treated Gestational Diabetes Mellitus. Diabetes Care 2018;41:1385–1390.

Fraser A, Nelson SM, Macdonald-Wallis C, Sattar N, Lawlor DA. Hypertensive disorders of pregnancy and cardiometabolic health in adolescent offspring. Hypertension 2013;62:614–620.

Zheng W, Zhang X, Feng Y, et al. Association of Corpus Callosum Development With Fetal Growth Restriction and Maternal Preeclampsia or Gestational Hypertension. JAMA Netw Open 2022;5:e2226696.

Ayano G, Betts K, Tait R, Dachew BA, Alati R. Maternal depressive and anxiety symptoms and the risk of attention deficit hyperactivity disorder symptoms in offspring aged 17:Findings from the Raine Study. J Affect Disord 2021;284:149–156.

Zhu Q, Yang X, Zhang Y, Shan C, Shi Z. Role of the Gut Microbiota in the Increased Infant Body Mass Index Induced by Gestational Diabetes Mellitus. mSystems 2022;7:e0046522.

Chu AHY, Godfrey KM. Gestational Diabetes Mellitus and Developmental Programming. Ann Nutr Metab 2020;76 Suppl 3:4-15.

Furse S, Koulman A, Ozanne SE, Poston L, White SL, Meek CL. Altered Lipid Metabolism in Obese Women With Gestational Diabetes and Associations With Offspring Adiposity. J Clin Endocrinol Metab 2022;107:e2825-e2832.

Kim C, Harrall KK, Glueck DH, Needham BL, Dabelea D. Gestational diabetes mellitus, epigenetic age and offspring metabolism. Diabet Med 2022;39:e14925.

Yu Y, Arah OA, Liew Z, et al. Maternal diabetes during pregnancy and early onset of cardiovascular disease in offspring:population based cohort study with 40 years of follow-up. BMJ 2019;367:l6398.

Yamamoto JM, Benham JL, Dewey D, et al. Neurocognitive and behavioural outcomes in offspring exposed to maternal pre-existing diabetes:a systematic review and meta-analysis. Diabetologia 2019;62:1561-1574.

Smith CA. Effects of maternal under nutrition upon the newborn infant in Holland (1944-1945). J Pediatr 1947;30:229-243.

Sarli PM, Manousopoulou A, Efthymiou E, et al. Liver Proteome Profile of Growth Restricted and Appropriately Grown Newborn Wistar Rats Associated With Maternal Undernutrition. Front Endocrinol (Lausanne) 2021;12:684220.

Zouridis A, Manousopoulou A, Potiris A, et al. Impact of Maternal Food Restriction on Heart Proteome in Appropriately Grown and Growth-Restricted Wistar-Rat Offspring. Nutrients 2021;13:466.

Potiris A, Manousopoulou A, Zouridis A, et al. The Effect of Prenatal Food Restriction on Brain Proteome in Appropriately Grown and Growth Restricted Male Wistar Rats. Front Neurosci 2021;15:665354.

Hagstrom H, Simon TG, Roelstraete B, Stephansson O, Soderling J, Ludvigsson JF. Maternal obesity increases the risk and severity of NAFLD in offspring. J Hepatol 2021;75:1042-1048.

Liu X, Li X, Xia B, et al. High-fiber diet mitigates maternal obesity-induced cognitive and social dysfunction in the offspring via gut-brain axis. Cell Metab 2021;33:923-938 e926.

Davenport BN, Wilson RL, Jones HN. Interventions for placental insufficiency and fetal growth restriction. Placenta 2022;125:4-9.

Eriksson JG, Kajantie E, Thornburg KL, Osmond C, Barker DJ. Mother's body size and placental size predict coronary heart disease in men. Eur Heart J 2011;32:2297-2303.

Hu C, Wu Z, Huang Z, Hao X, Wang S, Deng J. et al. Nox2 impairs VEGF-A-induced angiogenesis in placenta via mitochondrial ROS-STAT3 pathway. Redox Biol 2021;45:102051.

McIntyre KR, Hayward CE, Sibley CP, Greenwood SL, Dilworth MR. Evidence of adaptation of maternofetal transport of glutamine relative to placental size in normal mice, and in those with fetal growth restriction. J Physiol 2019;597:4975-4990.

Janzen C, Lei MY, Cho J, Sullivan P, Shin BC, Devaskar SU. Placental glucose transporter 3 (GLUT3) is up-regulated in human pregnancies complicated by late-onset intrauterine growth restriction. Placenta 2013;34:1072-1078.

Xu P, Wu Z, Xi Y, Wang L. Epigenetic regulation of placental glucose transporters mediates maternal cadmium-induced fetal growth restriction. Toxicology 2016;372:34-41.

Jansson T, Castillo-Castrejon M, Gupta MB, Powell TL, Rosario FJ. Down-regulation of placental Cdc42 and Rac1 links mTORC2 inhibition to decreased trophoblast amino acid transport in human intrauterine growth restriction. Clin Sci (Lond) 2020;134:53-70.

Chen YY, Rosario FJ, Shehab MA, Powell TL, Gupta MB, Jansson T. Increased ubiquitination and reduced plasma membrane trafficking of placental amino acid transporter SNAT-2 in human IUGR. Clin Sci (Lond) 2015;129:1131-1141.

Paolini CL, Marconi AM, Ronzoni S, et al. Placental transport of leucine, phenylalanine, glycine, and proline in intrauterine growth-restricted pregnancies. J Clin Endocrinol Metab 2001;86:5427-5432.

Johansson M, Karlsson L, Wennergren M, Jansson T, Powell TL. Activity and protein expression of Na+/K+ ATPase are reduced in microvillous syncytiotrophoblast plasma membranes isolated from pregnancies complicated by intrauterine growth restriction. J Clin Endocrinol Metab 2003;88:2831-2837.

Chassen SS, Ferchaud-Roucher V, Gupta MB, Jansson T, Powell TL. Alterations in placental long chain polyunsaturated fatty acid metabolism in human intrauterine growth restriction. Clin Sci (Lond) 2018;132:595-607.

Assumpcao RP, Mucci DB, Fonseca FCP, et al. Fatty acid profile of maternal and fetal erythrocytes and placental expression of fatty acid transport proteins in normal and intrauterine growth restriction pregnancies. Prostaglandins Leukot Essent Fatty Acids 2017;125:24-31.

Loubiere LS, Vasilopoulou E, Bulmer JN, et al. Expression of thyroid hormone transporters in the human placenta and changes associated with intrauterine growth restriction. Placenta 2010;31:295-304.

Chan SY, Franklyn JA, Pemberton HN, et al. Monocarboxylate transporter 8 expression in the human placenta:the effects of severe intrauterine growth restriction. J Endocrinol 2006;189:465-471.

Vasilopoulou E, Loubiere LS, Martin-Santos A, et al. Differential triiodothyronine responsiveness and transport by human cytotrophoblasts from normal and growth-restricted pregnancies. J Clin Endocrinol Metab 2010;95:4762-4770.

Settle P, Sibley CP, Doughty IM, et al. Placental lactate transporter activity and expression in intrauterine growth restriction. J Soc Gynecol Investig 2006;13:357-363.

Harrison DJ, Creeth HDJ, Tyson HR, et al. Placental endocrine insufficiency programs anxiety, deficits in cognition and atypical social behaviour in offspring. Hum Mol Genet 2021;30:1863-1880.

Whitehead CL, McNamara H, Walker SP, et al. Identifying late-onset fetal growth restriction by measuring circulating placental RNA in the maternal blood at 28 weeks' gestation. Am J Obstet Gynecol 2016;214:521 e521-521 e528.

Whitehead CL, Walker SP, Mendis S, Lappas M, Tong S. Quantifying mRNA coding growth genes in the maternal circulation to detect fetal growth restriction. Am J Obstet Gynecol 2013;209:133 e131-139.

Vacher CM, Lacaille H, O'Reilly JJ, et al. Placental endocrine function shapes cerebellar development and social behavior. Nat Neurosci 2021;24:1392-1401.

Sato K. Placenta-derived hypo-serotonin situations in the developing forebrain cause autism. Med Hypotheses 2013;80:368-372.

Ranzil S, Ellery S, Walker DW, et al. Disrupted placental serotonin synthetic pathway and increased placental serotonin:Potential implications in the pathogenesis of human fetal growth restriction. Placenta 2019;84:74-83.

Li D, Gao R, Qin L, Yue H, Sang N. New Insights into Prenatal NO2 Exposure and Behavioral Abnormalities in Male Offspring:Disturbed Serotonin Metabolism and Delayed Oligodendrocyte Development. Environ Sci Technol 2022;56:11536-11546.

Noorlander CW, Ververs FF, Nikkels PG, van Echteld CJ, Visser GH, Smidt MP. Modulation of serotonin transporter function during fetal development causes dilated heart cardiomyopathy and lifelong behavioral abnormalities. PLoS One 2008;3:e2782.

Wu YM, Luo HW, Kou H, et al. Prenatal caffeine exposure induced a lower level of fetal blood leptin mainly via placental mechanism. Toxicol Appl Pharmacol 2015;289:109-116.

Allbrand M, Eklund D, Cao Y, Nilsson K, Lodefalk M. Gene expression of leptin, leptin receptor isoforms and inflammatory cytokines in placentas of obese women- Associations to birth weight and fetal sex. Placenta 2022;117:64-71.

Visan V, Balan RA, Costea CF, et al. Morphological and histopathological changes in placentas of pregnancies with intrauterine growth restriction. Rom J Morphol Embryol 2020;61:477-483.

Wang T, Chen M, Yan YE, Xiao FQ, Pan XL, Wang H. Growth retardation of fetal rats exposed to nicotine in utero:possible involvement of CYP1A1, CYP2E1, and P-glycoprotein. Environ Toxicol 2009;24:33-42.

Ge C, Xu D, Yu P, et al. P-gp expression inhibition mediates placental glucocorticoid barrier opening and fetal weight loss. BMC Med 2021;19:311.

Marsit CJ, Maccani MA, Padbury JF, Lester BM. Placental 11-beta hydroxysteroid dehydrogenase methylation is associated with newborn growth and a measure of neurobehavioral outcome. PLoS One 2012;7:e33794.

Saegusa H, Nakagawa Y, Liu YJ, Ohzeki T. Influence of placental 11beta-hydroxysteroid dehydrogenase (11beta-HSD) inhibition on glucose metabolism and 11beta-HSD regulation in adult offspring of rats. Metabolism 1999;48:1584-1588.

Welberg LA, Seckl JR, Holmes MC. Inhibition of 11beta-hydroxysteroid dehydrogenase, the foeto-placental barrier to maternal glucocorticoids, permanently programs amygdala GR mRNA expression and anxiety-like behaviour in the offspring. Eur J Neurosci 2000;12:1047-1054.

Kratimenos P, Penn AA. Placental programming of neuropsychiatric disease. Pediatr Res 2019;86:157-164.

Behura SK, Kelleher AM, Spencer TE. Evidence for functional interactions between the placenta and brain in pregnant mice. FASEB J 2019;33:4261-4272.

Redline RW. Severe fetal placental vascular lesions in term infants with neurologic impairment. Am J Obstet Gynecol 2005;192:452-457.

Redline RW, O'Riordan MA. Placental lesions associated with cerebral palsy and neurologic impairment following term birth. Arch Pathol Lab Med 2000;124:1785-1791.

Whitehead E, Dodds L, Joseph KS, et al. Relation of pregnancy and neonatal factors to subsequent development of childhood epilepsy:a population-based cohort study. Pediatrics 2006;117:1298-1306.

Ruiz-Palacios M, Prieto-Sanchez MT, Ruiz-Alcaraz AJ, et al. Insulin Treatment May Alter Fatty Acid Carriers in Placentas from Gestational Diabetes Subjects. Int J Mol Sci 2017;18:1203.

Xu P, Guo H, Wang H, et al. Downregulations of placental fatty acid transporters during cadmium-induced fetal growth restriction. Toxicology 2019;423:112-122.

Goeden N, Velasquez J, Arnold KA, et al. Maternal Inflammation Disrupts Fetal Neurodevelopment via Increased Placental Output of Serotonin to the Fetal Brain. J Neurosci 2016;36:6041-6049.

Camm EJ, Botting KJ, Sferruzzi-Perri AN. Near to One's Heart:The Intimate Relationship Between the Placenta and Fetal Heart. Front Physiol 2018;9:629.

Barker DJ, Thornburg KL, Osmond C, Kajantie E, Eriksson JG. The surface area of the placenta and hypertension in the offspring in later life. Int J Dev Biol 2010;54:525-530.

Barker DJ, Gelow J, Thornburg K, Osmond C, Kajantie E, Eriksson JG. The early origins of chronic heart failure:impaired placental growth and initiation of insulin resistance in childhood. Eur J Heart Fail 2010;12:819-825.

Martyn CN, Barker DJ, Osmond C. Mothers' pelvic size, fetal growth, and death from stroke and coronary heart disease in men in the UK. Lancet 1996;348:1264-1268.

Burton GJ, Jauniaux E. Pathophysiology of placental-derived fetal growth restriction. Am J Obstet Gynecol 2018;218:S745-S761.

Linask KK, Han M, Bravo-Valenzuela NJ. Changes in vitelline and utero-placental hemodynamics:implications for cardiovascular development. Front Physiol 2014;5:390.

Maslen CL. Recent Advances in Placenta-Heart Interactions. Front Physiol 2018;9:735.

Linask KK. The heart-placenta axis in the first month of pregnancy:induction and prevention of cardiovascular birth defects. J Pregnancy 2013;2013:320413.

Kaufmann P, Mayhew TM, Charnock-Jones DS. Aspects of human fetoplacental vasculogenesis and angiogenesis. II. Changes during normal pregnancy. Placenta 2004;25:114-126.

Courtney JA, Cnota JF, Jones HN. The Role of Abnormal Placentation in Congenital Heart Disease;Cause, Correlate, or Consequence? Front Physiol 2018;9:1045.

Wilson RL, Yuan V, Courtney JA, Tipler A, Cnota JF, Jones HN. Analysis of commonly expressed genes between first trimester fetal heart and placenta cell types in the context of congenital heart disease. Sci Rep 2022;12:10756.

Woolford SJ, Curtis EM, D'Angelo S, et al. Placental volume at 11 weeks is associated with offspring bone mass at birth and in later childhood:Findings from the Southampton Women's Survey. Placenta 2020;99:101-107.

Holroyd CR, Osmond C, Barker DJ, et al. Placental Size Is Associated Differentially With Postnatal Bone Size and Density. J Bone Miner Res 2016;31:1855-1864.

Holroyd CR, Harvey NC, Crozier SR, et al. Placental size at 19 weeks predicts offspring bone mass at birth:findings from the Southampton Women's Survey. Placenta 2012;33:623-629.

Li SH, Yin HB, Ren MR, et al. TRPV5 and TRPV6 are expressed in placenta and bone tissues during pregnancy in mice. Biotech Histochem 2019;94:244-251.

Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS 2010;5:463-466.

Napso T, Zhao X, Lligona MI, et al. Placental secretome characterization identifies candidates for pregnancy complications. Commun Biol 2021;4:701.

Zamarian AC, Araujo Junior E, Daher S, Rolo LC, Moron AF, Nardozza LM. Evaluation of biochemical markers combined with uterine artery Doppler parameters in fetuses with growth restriction:a case-control study. Arch Gynecol Obstet 2016;294:715-723.

Yu N, Cui H, Chen X, Chang Y. First trimester maternal serum analytes and second trimester uterine artery Doppler in the prediction of preeclampsia and fetal growth restriction. Taiwan J Obstet Gynecol 2017;56:358-361.

Chafetz I, Kuhnreich I, Sammar M, et al. First-trimester placental protein 13 screening for preeclampsia and intrauterine growth restriction. Am J Obstet Gynecol 2007;197:35 e31-37.

Lo YM, Corbetta N, Chamberlain PF, et al. Presence of fetal DNA in maternal plasma and serum. Lancet 1997;350:485-487.

Poon LL, Leung TN, Lau TK, Lo YM. Presence of fetal RNA in maternal plasma. Clin Chem 2000;46:1832-1834.

Tsui NB, Chim SS, Chiu RW, et al. Systematic micro-array based identification of placental mRNA in maternal plasma:towards non-invasive prenatal gene expression profiling. J Med Genet 2004;41:461-467.

Go AT, van Vugt JM, Oudejans CB. Non-invasive aneuploidy detection using free fetal DNA and RNA in maternal plasma:recent progress and future possibilities. Hum Reprod Update 2011;17:372-382.

Chim SS, Shing TK, Hung EC, et al. Detection and characterization of placental microRNAs in maternal plasma. Clin Chem 2008;54:482-490.

Jelena M, Sopic M, Joksic I, et al. Placenta-specific plasma miR518b is a potential biomarker for preeclampsia. Clin Biochem 2020;79:28-33.

Vlahos A, Mansell T, Saffery R, Novakovic B. Human placental methylome in the interplay of adverse placental health, environmental exposure, and pregnancy outcome. PLoS Genet 2019;15:e1008236.

Ravaei A, Emanuele M, Nazzaro G, Fadiga L, Rubini M. Placental DNA methylation profile as predicting marker for autism spectrum disorder (ASD). Mol Med 2023;29:8.

Schroeder DI, Schmidt RJ, Crary-Dooley FK, et al. Placental methylome analysis from a prospective autism study. Mol Autism 2016;7:51.

Zhu Y, Mordaunt CE, Yasui DH, et al. Placental DNA methylation levels at CYP2E1 and IRS2 are associated with child outcome in a prospective autism study. Hum Mol Genet 2019;28:2659-2674.

Radhakrishna U, Albayrak S, Zafra R, et al. Placental epigenetics for evaluation of fetal congenital heart defects:Ventricular Septal Defect (VSD). PLoS One 2019;14:e0200229.

Paauw ND, Lely AT, Joles JA, et al. H3K27 acetylation and gene expression analysis reveals differences in placental chromatin activity in fetal growth restriction. Clin Epigenetics 2018;10:85.

Pedroso MA, Palmer KR, Hodges RJ, Costa FDS, Rolnik DL. Uterine Artery Doppler in Screening for Preeclampsia and Fetal Growth Restriction. Rev Bras Ginecol Obstet 2018;40:287-293.

Bakalis S, Peeva G, Gonzalez R, Poon LC, Nicolaides KH. Prediction of small-for-gestational-age neonates:screening by biophysical and biochemical markers at 30-34 weeks. Ultrasound Obstet Gynecol 2015;46:446-451.

Dieste-Perez P, Saviron-Cornudella R, Tajada-Duaso M, et al. Personalized Model to Predict Small for Gestational Age at Delivery Using Fetal Biometrics, Maternal Characteristics, and Pregnancy Biomarkers:A Retrospective Cohort Study of Births Assisted at a Spanish Hospital. J Pers Med 2022;12:762.

Kienast C, Moya W, Rodriguez O, Jijon A, Geipel A. Predictive value of angiogenic factors, clinical risk factors and uterine artery Doppler for pre-eclampsia and fetal growth restriction in second and third trimester pregnancies in an Ecuadorian population. J Matern Fetal Neonatal Med 2016;29:537-543.

Kwiatkowski S, Bednarek-Jedrzejek M, Ksel J, et al. sFlt-1/PlGF and Doppler ultrasound parameters in SGA pregnancies with confirmed neonatal birth weight below 10th percentile. Pregnancy Hypertens 2018;14:79-85.

Garcia-Manau P, Mendoza M, Bonacina E, et al. Soluble fms-like tyrosine kinase to placental growth factor ratio in different stages of early-onset fetal growth restriction and small for gestational age. Acta Obstet Gynecol Scand 2021;100:119-128.

Chen W, Wei Q, Liang Q, Song S, Li J. Diagnostic capacity of sFlt-1/PlGF ratio in fetal growth restriction:A systematic review and meta-analysis. Placenta 2022;127:37-42.

Cowans NJ, Spencer K. First-trimester ADAM12 and PAPP-A as markers for intrauterine fetal growth restriction through their roles in the insulin-like growth factor system. Prenat Diagn 2007;27:264-271.

Stepan H, Kramer T, Faber R. Maternal plasma concentrations of soluble endoglin in pregnancies with intrauterine growth restriction. J Clin Endocrinol Metab 2007;92:2831-2834.

Asvold BO, Vatten LJ, Romundstad PR, Jenum PA, Karumanchi SA, Eskild A. Angiogenic factors in maternal circulation and the risk of severe fetal growth restriction. Am J Epidemiol 2011;173:630-639.

Nanjo S, Minami S, Mizoguchi M, et al. Levels of serum-circulating angiogenic factors within 1 week prior to delivery are closely related to conditions of pregnant women with pre-eclampsia, gestational hypertension, and/or fetal growth restriction. J Obstet Gynaecol Res 2017;43:1805-1814.

Li Y, Liu J. MicroRNA-206 predicts raised fetal growth retardation risk through the interaction with vascular endothelial growth factor in pregnancies. Medicine (Baltimore) 2020;99:e18897.

Tang Y, Ye W, Liu X, Lv Y, Yao C, Wei J. VEGF and sFLT-1 in serum of PIH patients and effects on the foetus. Exp Ther Med 2019;17:2123-2128.

Bourque DK, Avila L, Penaherrera M, von Dadelszen P, Robinson WP. Decreased placental methylation at the H19/IGF2 imprinting control region is associated with normotensive intrauterine growth restriction but not preeclampsia. Placenta 2010;31:197-202.

Qiu Q, Bell M, Lu X, et al. Significance of IGFBP-4 in the development of fetal growth restriction. J Clin Endocrinol Metab 2012;97:E1429-1439.

Pang WW, Tsui MH, Sahota D, et al. A strategy for identifying circulating placental RNA markers for fetal growth assessment. Prenat Diagn 2009;29:495-504.

Kaitu'u-Lino TJ, MacDonald TM, Cannon P, et al. Circulating SPINT1 is a biomarker of pregnancies with poor placental function and fetal growth restriction. Nat Commun 2020;11:2411.

Kaitu'u-Lino TJ, Tong S, Walker SP, et al. Maternal circulating SPINT1 is reduced in small-for-gestational age pregnancies at 26 weeks:Growing up in Singapore towards health outcomes (GUSTO) cohort study. Placenta 2021;110:24-28.

Murphy CN, Cluver CA, Walker SP, et al. Circulating SPINT1 Is Reduced in a Preeclamptic Cohort with Co-Existing Fetal Growth Restriction. J Clin Med 2022;11:901.

MacDonald TM, Walker SP, Hiscock R, Cannon P, Harper A, Murray E. et al. Circulating Delta-like homolog 1 (DLK1) at 36 weeks is correlated with birthweight and is of placental origin. Placenta 2020;91:24-30.

Krauss T, Emons G, Kuhn W, Augustin HG. Predictive value of routine circulating soluble endothelial cell adhesion molecule measurements during pregnancy. Clin Chem 2002;48:1418-1425.

Schneuer FJ, Nassar N, Khambalia AZ, et al. First trimester screening of maternal placental protein 13 for predicting preeclampsia and small for gestational age:in-house study and systematic review. Placenta 2012;33:735-740.

Spencer K, Yu CK, Cowans NJ, Otigbah C, Nicolaides KH. Prediction of pregnancy complications by first-trimester maternal serum PAPP-A and free beta-hCG and with second-trimester uterine artery Doppler. Prenat Diagn 2005;25:949-953.

Gundu S, Kulkarni M, Gupte S, Gupte A, Gambhir M, Gambhir P. Correlation of first-trimester serum levels of pregnancy-associated plasma protein A with small-for-gestational-age neonates and preterm births. Int J Gynaecol Obstet 2016;133:159-163.

Canini S, Prefumo F, Pastorino D, et al. Association between birth weight and first-trimester free beta-human chorionic gonadotropin and pregnancy-associated plasma protein A. Fertil Steril 2008;89:174-178.

Crovetto F, Crispi F, Scazzocchio E, et al. First-trimester screening for early and late small-for-gestational-age neonates using maternal serum biochemistry, blood pressure and uterine artery Doppler. Ultrasound Obstet Gynecol 2014;43:34-40.

Erez O, Romero R, Espinoza J, et al. The change in concentrations of angiogenic and anti-angiogenic factors in maternal plasma between the first and second trimesters in risk assessment for the subsequent development of preeclampsia and small-for-gestational age. J Matern Fetal Neonatal Med 2008;21:279-287.

Regnault TR, Orbus RJ, de Vrijer B, et al. Placental expression of VEGF, PlGF and their receptors in a model of placental insufficiency-intrauterine growth restriction (PI-IUGR). Placenta 2002;23:132-144.

Sifakis S, Akolekar R, Kappou D, Mantas N, Nicolaides KH. Maternal serum placental growth hormone at 11-13 weeks' gestation in pregnancies delivering small for gestational age neonates. J Matern Fetal Neonatal Med 2012;25:1796-1799.

Cleaton MA, Dent CL, Howard M, et al. Fetus-derived DLK1 is required for maternal metabolic adaptations to pregnancy and is associated with fetal growth restriction. Nat Genet 2016;48:1473-1480.

Pham A, Mitanchez D, Forhan A, et al. Low Maternal DLK1 Levels at 26 Weeks Is Associated With Small for Gestational Age at Birth. Front Endocrinol (Lausanne) 2022;13:836731.

Ashraf UM, Hall DL, Rawls AZ, Alexander BT. Epigenetic processes during preeclampsia and effects on fetal development and chronic health. Clin Sci (Lond) 2021;135:2307-2327.

Stepan H, Galindo A, Hund M, et al. Clinical utility of sFlt-1 and PlGF in screening, prediction, diagnosis and monitoring of pre-eclampsia and fetal growth restriction. Ultrasound Obstet Gynecol 2023;61:168-180.

Guo Z, Yang F, Zhang J, et al. Whole-Genome Promoter Profiling of Plasma DNA Exhibits Diagnostic Value for Placenta-Origin Pregnancy Complications. Adv Sci (Weinh) 2020;7:1901819.

Bahado-Singh R, Vishweswaraiah S, Mishra NK, Guda C, Radhakrishna U. Placental DNA methylation changes in detection of tetralogy of Fallot. Ultrasound Obstet Gynecol 2020;55:768-775.

Liu J, Wu Y, Sun H, et al. Association between placental DNA methylation and fetal congenital heart disease. Mol Genet Genomics 2023;298:243-251.

Liu J, Wu Y, Sun H, et al. Placental DNA Methylation Abnormalities in Prenatal Conotruncal Heart Defects. Front Genet 2022;13:878063.

Zhu Y, Gomez JA, Laufer BI, Mordaunt CE, Mouat JS, Soto DC. et al. Placental methylome reveals a 22q13.33 brain regulatory gene locus associated with autism. Genome Biol 2022;23:46.

Bahado-Singh RO, Vishweswaraiah S, Aydas B, Radhakrishna U. Placental DNA methylation changes and the early prediction of autism in full-term newborns. PLoS One 2021;16:e0253340.

Appleton AA, Lester BM, Armstrong DA, Lesseur C, Marsit CJ. Examining the joint contribution of placental NR3C1 and HSD11B2 methylation for infant neurobehavior. Psychoneuroendocrinology 2015;52:32-42.

Ursini G, Di Carlo P, Mukherjee S, et al. Prioritization of potential causative genes for schizophrenia in placenta. Nat Commun 2023;14:2613.

Tehrani JM, Kennedy E, Tung PW, et al. Human placental microRNAs dysregulated by cadmium exposure predict neurobehavioral outcomes at birth. Pediatr Res 2023;93:1410-1418.

Howerton CL, Morgan CP, Fischer DB, Bale TL. O-GlcNAc transferase (OGT) as a placental biomarker of maternal stress and reprogramming of CNS gene transcription in development. Proc Natl Acad Sci U S A 2013;110:5169-5174.

Nugent BM, O'Donnell CM, Epperson CN, Bale TL. Placental H3K27me3 establishes female resilience to prenatal insults. Nat Commun 2018;9:2555.

Chhabra D, Sharma S, Kho AT, et al. Fetal lung and placental methylation is associated with in utero nicotine exposure. Epigenetics 2014;9:1473-1484.

Shorey-Kendrick LE, McEvoy CT, O'Sullivan SM, et al. Impact of vitamin C supplementation on placental DNA methylation changes related to maternal smoking:association with gene expression and respiratory outcomes. Clin Epigenetics 2021;13:177.

Arendt LH, Ramlau-Hansen CH, Wilcox AJ, Henriksen TB, Olsen J, Lindhard MS. Placental Weight and Male Genital Anomalies:A Nationwide Danish Cohort Study. Am J Epidemiol 2016;183:1122-1128.

Arrebola JP, Molina-Molina JM, Fernandez MF, et al. A novel biomarker for anti-androgenic activity in placenta reveals risks of urogenital malformations. Reproduction 2015;149:605-613.

Fernandez MF, Olmos B, Granada A, et al. Human exposure to endocrine-disrupting chemicals and prenatal risk factors for cryptorchidism and hypospadias:a nested case-control study. Environ Health Perspect 2007;115:8-14.

Green BB, Kappil M, Lambertini L, et al. Expression of imprinted genes in placenta is associated with infant neurobehavioral development. Epigenetics 2015;10:834-841.

Lesseur C, Armstrong DA, Murphy MA, et al. Sex-specific associations between placental leptin promoter DNA methylation and infant neurobehavior. Psychoneuroendocrinology 2014;40:1-9.

Redline RW, Minich N, Taylor HG, Hack M. Placental lesions as predictors of cerebral palsy and abnormal neurocognitive function at school age in extremely low birth weight infants (<1 kg). Pediatr Dev Pathol 2007;10:282-292.

Maccani MA, Padbury JF, Lester BM, Knopik VS, Marsit CJ. Placental miRNA expression profiles are associated with measures of infant neurobehavioral outcomes. Pediatr Res 2013;74:272-278.

Goodfellow LR, Cooper C, Harvey NC. Regulation of placental calcium transport and offspring bone health. Front Endocrinol (Lausanne) 2011;2:3.

Published

2024-03-14

How to Cite

1.
Yu P, Chen S, Wang H. Placental biomarkers of fetal-originated diseases. PRM. 2024;3. doi:10.54844/prm.2024.0466

Issue

Section

Review Article

Downloads

Download data is not yet available.
REVIEW ARTICLE

Placental biomarkers of fetal-originated diseases


Pengxia Yu1, Sijia Chen2,3, Hui Wang2,3,*

1Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, Hubei Province, China

2Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, Hubei Province, China

3Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, Hubei Province, China


*Corresponding Author:

Hui Wang, Department of Pharmacology, Basic Medical School of Wuhan University, 185 Donghu Road, Wuchang District, Wuhan 430071, Hubei Province, China. E-mail: wanghui19@whu.edu.cn; ORCID: https://orcid.org/0000-0001-5300-8661


Received: 21 September 2023 Revised: 6 February 2024 Accepted: 27 February 2024


ABSTRACT

Fetal-originated diseases refer to birth defects of offspring and multiple chronic diseases in adulthood caused by abnormal embryonic development. However, due to the vulnerability of fetuses and newborns, as well as technical limitations, it is very difficult to carry out effective development assessment and early warning of long-term diseases in the early stages of life. It is known that the placenta is the unique link between the mother and the fetus, and its role in the occurrence and progression of fetal-originated diseases cannot be ignored. Studies have found that a variety of adverse environmental factors (such as nanoparticle exposure) cannot pass through the placental barrier, but can lead to fetal dysplasia and multi-organ development programming changes by affecting placental development, and ultimately mediate the occurrence of fetal-originated diseases. Meanwhile, some environmental factors that can pass through the placental barrier can cause placenta-fetal co-exposure, resulting in similar signaling pathways and epigenetic changes. The placenta originates from the fetus and the mother, and its development is accompanied by changes in indicators that can be objectively and quantitatively detected. These factors can be used as a biomarker to assess maternal exposure, and placental function, and to predict the developmental status and long-term disease susceptibility of offspring. To date, researchers have discovered a variety of potential placental biomarkers, and show promising application prospects. This paper reviews the recent research on placenta-related mechanisms leading to fetal-originated diseases and placental biomarkers, to provide the theoretical and experimental basis for early warning, prevention, and treatment of fetal-originated diseases.

Key words: placenta, adverse environment, fetal growth restriction, fetal-originated diseases, biomarkers

INTRODUCTION

The "Developmental Origins of Health and Disease (DOHaD)" theory proposes that experiencing adverse environments early in life increases the incidence of chronic diseases such as obesity, diabetes, and cardiovascular disease in adulthood.[1] Despite the developmental plasticity of fetal organs and systems, a variety of adverse environments can lead to irreversible changes in fetal development programming, laying the groundwork for adverse pregnancy outcomes and long-term disease susceptibility.[2] The placenta is the unique interface connecting the mother and the fetus. Functionally, the placenta sustains the growth of the fetus as it facilitates the delivery of oxygen and nutrients and the removal of waste products. Meanwhile, the placenta is an active endocrine organ, secreting a plethora of factors, such as steroids, glycoproteins, peptide hormones, cytokines, and neuroactive factors, that prevent immune rejection of the fetus and regulate the maternal pregnancy process and fetal development. In addition, the placenta is also a physical and biological barrier between the mother and the fetal circulatory system, which can protect the fetus from adverse environmental interference. When the placenta has abnormal morphology and functional damage, it is often accompanied by objectively and quantitatively detectable biochemical molecules, signal transducers, functional proteins, and epigenetic modification changes. These indicators can be used as placental biomarkers to evaluate placental function and fetal development, and even have the potential to predict the occurrence and progression of fetal diseases after birth. At present, the combination of magnetic resonance examination of placenta structure, placental biomarkers, and ultrasonic fetal bioassay has shown a strong ability to predict the occurrence of fetal growth restriction (FGR) and related complications. Meanwhile, the development of "multi-omics" technology offers great possibilities for the identification of novel biomarkers related to fetal growth. This review will discuss the placental origin of fetal-originated diseases and summarize the latest research on placental biomarkers to provide the theoretical basis for early warning and comprehensive prevention and treatment of fetal-originated diseases.

THE ADVERSE ENVIRONMENT DURING PREGNANCY LEADS TO THE OCCURRENCE OF FETAL-ORIGINATED DISEASES

The fetal-originated diseases refer to birth defects caused by abnormal embryonic development and a variety of chronic diseases in adulthood, such as diabetes, fatty liver, and osteoporosis.[3] In addition to genetic factors, the occurrence of fetal-originated diseases is largely affected by adverse environments and is closely related to altered programming of fetal development. The adverse environment during pregnancy can be divided into exogenous factors and maternal factors. It has been found that most of the adverse environmental factors can cross the placental barrier and directly affect embryonic or fetal development, such as polycyclic aromatic hydrocarbons,[4] particulate matter 2.5 (PM2.5),[5,6] dexamethasone,[7,8] heavy metals,[9,10] nicotine,[11,12] ethanol,[13] caffeine,[14,15] cocaine,[16,17] and opioids,[1719]etc. Besides, some environmental factors that cannot cross the placental barrier can indirectly affect fetal development by disrupting the structure and function of the placenta, such as exposure to cobalt and chromium nanoparticles causing placental autophagic flow impairment and interleukin (IL)-6 release, which disrupts fetal astrocytes and neurons development.[20] This review lists some common adverse environmental factors and their effects on the early development and long-term health of offspring (Table 1). In summary, the adverse environment during pregnancy can lead to adverse pregnancy outcomes such as FGR and increased susceptibility to a variety of chronic diseases after birth by interfering with the developmental programming of multiple organs.

Table 1: Adverse environment during pregnancy and effects on early development and long-term health of offspring
Adverse environment type Designation Cross placental barrier Effects on offspring development Species References
Environmental toxic substance PAH Yes FGR; Behavior disorder, allergies, or asthma Human [21,22]
1-NP Yes Interference with placental blood vessels and proliferation results in fetal growth restriction Mice [23]
PBDE Yes FGR, Changes in placental methylation Human [24,25]
DEHP Yes Interference with placental thyroid hormone receptor signaling results in FGR; Impaired male reproductive development and neurodevelopment Mice [26]
Fenvalerate Yes Oxidative stress damage to the placenta; FGR Mice [27,28]
Cadmium Yes Inhibits placental progesterone synthesis.
Mitochondrial autophagy in placental trophoblast cells; FGR; Fetal liver and lung weight reduced
Mice [9,10]
Mercury Yes FGR; Anxiety-like behavior; Altered glucose metabolism; Insulin resistance Human; Mice [29,30]
PM2.5 Yes Low birth weight; cardiovascular disease in adults Mice; Human [5,6]
Bisphenol A Yes FGR; Spiral artery remodeling; Testicular tumor Mice [31,32]
Pesticide Yes Placental maturity and FGR Human [33]
Phthalate Yes FGR Human [34]
Tobacco smoke Yes FGR Human [35]
Nicotine Yes FGR; The programmed changes of neuroendocrine metabolism in offspring rats; Fetal metabolomics changes Rat [11,12]
Nanoparticles (cobalt and chromium) No Abnormal fetal neurodevelopment Human; Mice [20,36]
Drugs Benzedrine Yes Preeclampsia, placental abruption Human [37]
Dexamethasone Yes FGR; Multiple organ dysplasia Mice; Human [7,8]
Acetaminophen Yes Childhood asthma Human [38]
Cocaine Yes Spontaneous abortion; Stillbirth; Birth weight, head circumference, length, and Apgar score all decreased; Vascular dysfunction in adult offspring; Increased susceptibility to depressive and anxiety-like behavior, memory impairments, and epilepsy Human; Rat [3941]
Opiates Yes Neonatal abstinence syndrome; Low birth weight; Increased risk of spina bifida Human; [4244]
Marijuana Yes Low birth weight; Attention scores are worse in early childhood; Mid-childhood psychopathological symptoms
Glucose metabolism dysfunction; cardiovascular disease
Human [4547]
Methylamphetamine Yes Low birth weight; cardiovascular disease; Neurocognitive and behavioral deficits Mice; Rat; Human [4850]
Azithromycin No Low birth weight; Abnormal development of long bones, cartilage, liver, and gonads; Increased risk of spontaneous abortion Mice; Human [51,52]
Insulin No neonatal hypoglycemia; Human [53]
Food and beverage Alcohol Yes Fetal alcohol spectrum disorder Human [13]
Caffeine Yes FGR; Dysfunction of adrenal steroid synthesis Rat; Human [14,15]
maternal health factors Gestational hypertension/ Preeclampsia - FGR; Abnormal development of the fetal corpus callosum; Cardiovascular risk in children and young adults Human [54,55]
Depression - Attention deficit/hyperactivity disorder symptoms in childhood Human [56]
Gestational diabetes - Fetal overgrowth; Obesity; Lower insulin sensitivity; Cardiovascular risk from childhood to early adulthood; Neurocognitive and behavioral deficits Human [57,58] [57,5962]
Malnutrition - FGR; Hepatic metabolic changes; Cardiac development changes; Cognitive and neurodevelopmental impairments Human; Rat [6366]
Obesity - Non-alcoholic fatty liver disease; Neurodevelopmental disorders; Human [67,68]
PAH, polycyclic aromatic hydrocarbons; FGR, fetal growth restriction; 1-NP, 1-nitropyrene; PBDE, polybrominated diphenyl ethers; DEHP, diesters of phthalate; PM2.5, particulate matter 2.5.

PLACENTAL ORIGIN OF FETAL-ORIGINATED DISEASES

At present, an increasing number of clinical and animal experimental studies have led to a deeper understanding of the biological mechanism of the developmental origin of diseases. The placenta plays an important role in the occurrence and progression of fetal-originated diseases, which means that there is a placental origin for fetal-originated diseases. In this review, we focus on the important role of the placenta in fetal-originated diseases.

Placenta and fetal growth

Slowed fetal growth occurs in about 10% of pregnancies, but only a few have a pathological background known as FGR or intrauterine growth retardation (IUGR). Studies have found that approximately 75% of FGR is due to abnormal placental development.[69] Changes in the shape and size of the placenta lead to a decrease in the volume, efficiency, and function of the placenta. The Helsinki Cohort Study showed that placental size is associated with fetal FGR,[70] possibly due to vascular dysplasia in the low-weight placenta, which impairs the exchange of oxygen and nutrients between mother and fetus.[71] The placenta has a variety of functions, such as transport, endocrine, barrier, and so on, and acts as the lungs, intestines, liver, and kidneys of the fetus during pregnancy. The proper development of the placenta is essential to ensure fetal growth, and the occurrence of FGR is closely related to placental dysfunction. This review summarizes the abnormalities of placental function associated with fetal-originated diseases (Table 2).

Table 2: Fetal-originated diseases caused by placental dysfunction
Function Mechanism Pregnancy outcome and long-term disease Species References
Transport mTOR inhibition reduces glutamine transport FGR Mice [72]
GLUT3 upregulated FGR Human; Mice [73,74]
Inhibition of amino acid transport expression FGR Human [7577]
Down-regulation of Na+/K+-ATPase FGR Human [78]
Abnormal expression of fatty acid transporters FGR Human [79,80]
Abnormal expression of thyroid hormone transporters FGR Human [8183]
Decreased lactate transport capacity FGR Human [84]
Endocrine Placental endocrine insufficiency FGR; Anxiety-like behaviors, cognitive deficits, and atypical social behaviors increased Mice [85]
Abnormal secretion of IGF-1 FGR Human [86,87]
Lack of allopregnanolone Autism spectrum disorder-like behaviors in male offspring Mice [88]
Dysregulation of serotonin synthesis, metabolism, and transport in the placenta FGR; Autism spectrum disorder and anxiety- and depression-like behaviors Mice; Human [8992]
The short leptin receptor and leptin expression were decreased FGR Rat; Human [93,94]
Barrier fibrin deposits and diffuse calcification FGR Human [95]
The expression of P-gp decreased FGR Rat [96,97]
Increased methylation and decreased expression of 11β-HSD2 in the placenta FGR; Neonatal neurobehavioral changes; HPA axis changes, anxiety-like behavior, glucose intolerance in adult offspring Human; Rat [98100]
mTOR, mammalian target of rapamycin; FGR, fetal growth restriction; GLUT3, glucose transporter 3; IGF-1, insulin-like growth factor-1; P-gp, p-glycoprotein; 11β-HSD2, 11β-hydroxysteroid dehydrogenase 2; HPA, hypothalamic-pituitary-adrenal.

Placenta-fetal organ axis

The placenta originates from the trophectoderm of the blastocyst and shares the same genetic information and a similar developmental trajectory as the fetus. Some adverse factors that can cross the placental barrier, such as environmental pollution, endocrine disruptors, heavy metals, etc., can cause synchronous destruction of the common development pathways of the placental and fetus, resulting in abnormal placenta-fetal development and susceptibility to long-term diseases. As research progressed, a close link between the placenta and the development of certain fetal organs (e. g., the brain and heart) was demonstrated, giving rise to terms such as the "placenta-brain axis" and the "placenta-heart axis".

Placenta-brain axis

It has been reported that the placenta protects and shapes the fetal brain through the "placenta-brain axis", mainly through its gas exchange, nutrient transport, and endocrine functions.[101] A study analyzed the differentially regulated receptor-ligand gene expression patterns between the fetal brain and placenta in Foxa2 knockout mice and found that it is mainly associated with biological functions such as brain development, fetal response to oxygen, and regulation of synaptic transmission.[102] The placental vascular bed is the interface between the maternal and fetal and receives 25% to 32% of the total fetal cardiac output. Therefore, placental vascular abnormalities can have a significant impact on the overall circulatory function of the fetus, especially the blood flow to the central nervous system. Reports have shown that the occurrence, duration, and anatomical location of four types of severe placental fetal vasculopathy (including fetal thrombotic vasculopathy, chronic chorioamnionitis with occlusion fetal vasculopathy, chorioamnionitis with severe fetal vasculopathy, and meconium-associated fetal vascular necrosis) are highly correlated with neurological impairment and cerebral palsy.[103] It may be caused by poor perfusion of placental blood vessels, chronic inflammation, and hypoplasia of villi, which lead to impaired functional brain development by decreasing the oxygen delivery to the fetal brain.[103,104] In addition, Whitehead et al. reported a significant association between bleeding at the decidual-placental interface due to placental abruption and childhood epilepsy in a large population-based cohort study.[105] Fatty acids are important components of fetal brain adipose tissue, and n-3 and n-6 fatty acids can only be obtained from the maternal diet and transported to the fetal side through the placenta. Studies have found that long-chain fatty acid transport between mother and fetus is impaired in pregnancies associated with placental dysplasia (e. g., gestational diabetes mellitus, cadmium exposure) and affects fetal brain development and long-term health.[106,107] In addition to transferring nutrients and gases, the placenta produces a variety of neurotransmitters and hormones that can enter the fetal circulation and affect brain development, such as 5-hydroxy tryptamine (5-HT), dopamine, adrenaline/norepinephrine, and tetrahydroprogesterone, thereby affecting fetal neurogenesis and neuronal migration. It has been reported that maternal inflammation in the second trimester can lead to upregulation of 5-HT transformation in the mice placenta, inhibit the growth of endogenous 5-HT axons in the fetal forebrain, disrupt the fetal neurodevelopment process, which in turn affects adult brain functions, such as memory deficits.[108] This remarkable correlation between the placenta and the brain is known as the "placenta-brain axis" and has spurred the development of the field of placenta-neuro development.

Placenta-heart axis

The placenta is the "gatekeeper" of fetal heart development.[109] Cohort studies have shown a link between placental phenotypic characteristics and an increased risk of cardiovascular disease in adulthood.[70,110,111] Martyn et al. found that stroke and coronary heart disease mortality tended to be highest in British men with low birth weight,[112] which was one of the first studies to link placenta to cardiovascular disease. Subsequently, the intricate relationship between the placenta and the fetal heart was highlighted in clinical cases of placental pathology. In placental dysplasia associated with FGR, inadequate remodeling of the uterine spiral arteries leads to poor placental perfusion and increased vascular resistance, which can impair placental endocrine and transport function.[113] Approximately 45% of the ventricular output of the fetal heart flows directly to the placenta, and a higher placental vascular resistance will result in an increased fetal cardiac load on the fetus. Therefore, placental abnormalities may disrupt the development of the heart by altering the hemodynamics and leading to fetal death.[114] The development of the placenta and the fetal heart are parallel, and the two organs share several developmental pathways and also have the same susceptibility to genetic defects, known as the "placenta-heart axis".[115117] In early pregnancy, gene expression changes associated with angiogenesis and angiogenesis can lead to both placental and cardiac defects.[118] Currently, 329 genes have been identified through the mouse genome informatics database that can simultaneously cause placental morphology abnormalities and cardiovascular defects.[109] A single-cell sequencing analysis showed that cardiomyocytes, endothelial cells, and placental trophoblast cells share common expression genes and signaling pathways in early pregnancy. These genes and signaling pathways are essential for normal cell and organ function and have been implicated in coronary heart disease and placenta-related diseases.[119] For instance, the classical Wnt/β-catenin pathways are critical for placental development, implantation, and cardiac development in early pregnancy, and alterations in these pathways may lead to perturbations in the development of both organs simultaneously.[116]

Others

In addition to the placenta-brain axis and placenta-heart axis, there are also significant associations between the placenta and other tissues or organs, but due to the paucity of research, it is not widely recognized at present. For example, several population-based mother-child cohort studies have found that placental size is associated with skeletal development in the offspring,[120] and this association persists into childhood and even adolescence.[121,122] Transient receptor potential cation channel subfamily V member 5 (TRPV5) and TRPV6 are involved in calcium transport in the placenta and bone and can affect calcium homeostasis during both placental and fetal bone development.[123] In short, there may be interactions or similar changes and injuries between the placenta and various organs of the fetus in the intrauterine co-exposure environment, suggesting the existence of a "placenta-fetus organ axis".

PLACENTAL BIOMARKER TYPES

Biomarkers are objective measurements that allow the assessment of physiological activity, pathological progression, and pharmacological response.[124] Biomarkers are extremely valuable in both in vivo and in vitro experiments, as well as in the early efficacy and safety evaluation of clinical trials. The placenta produces a variety of important substances that enter the maternal circulation to maintain a normal pregnancy. In response to adverse environmental, significant molecular changes in the placenta may indicate specific fetal pathological features. Although there are still no placental biomarkers for predicting fetal-originated diseases for clinical use, detection of placenta-derived substances in maternal blood (such as bioactive peptides and subcellular fragments), or detection of placental gene expression patterns after delivery (such as epigenetic modifications) can be useful for indicating fetal multiple organ development abnormalities or predicting future susceptibility to fetal-originated diseases. Here we list some types of placental biomarkers that can be used to monitor placenta-fetal development in real time or to predict future disease occurrence in offspring.

Bioactive peptides

The placenta produces a variety of bioactive peptides that contribute to maternal adaptation to pregnancy. It has long been suggested that the detection of placenta-derived bioactive peptides in maternal blood may provide diagnostic or prognostic value for pregnancy complications. For example, the detection of placenta-secreted human chorionic gonadotropin (hCG) in maternal blood is used to confirm pregnancy, detection of placenta-derived angiogenesis regulator soluble fms-like tyrosine kinase 1 (sFLT1) and placental growth factor (PlGF) in maternal blood can be used to predict preeclampsia (PE). Some researchers have also combined molecular tracking techniques and bioinformatics analysis to integrate the secreted protein maps of human and mouse placentas, validating in clinical cases that detection of the relative abundance of secreted placenta proteins at 12 weeks of gestation can be used for the early diagnosis of pregnancy complications, and identified a variety of factors that may affect placental hormone expression and pregnancy outcome.[125] With the deepening of research, a variety of bioactive factors secreted by the placenta (Pregnancy-associated plasma protein A [PAPP-A], sEng, insulin-like growth factor [IGF], A disintegrin and metalloproteinase [ADAM12], Placental protein 13 [PP-13], etc.) are useful for monitoring fetal development and are expected to be early predictive biomarkers of FGR, although their applicability as clinical markers needs more verification.[126128]

Subcellular fractions

Several emerging technologies are being used to monitor or assess fetal development. For example, subcellular fractions derived from the placenta or fetus in the maternal circulation can be detected, including cell-free DNA (cfDNA), RNA, and miRNA. Studies as early as 1997 and 2000 have found the presence of fetal DNA and RNA in maternal blood.[129,130] Maternal blood cfDNA, derived primarily from placental trophoblasts and maternal hematopoietic cells, contains a nucleosome footprint that carries information about its origin tissue and gene expression, which allows for non-invasive monitoring of pregnancy-related complications and has the potential to predict adverse pregnancy outcomes. Placental mRNA in maternal plasma is a stable and easily detectable subcellular fraction. In 2004, Tsui et al. systematically evaluated the expression profiles of placental tissues by microarray analysis and demonstrated that placental gene expression levels exceeding a specific threshold can usually be detected in maternal plasm.[131] Some of these placental-specific mRNAs, including chorionic growth hormone 1, placental growth hormone 2 (PGH2), kisspeptin-1 (KISS1), and A disintegrin and metalloprotease (ADAM-12) are functionally associated with fetal or placental development. Non-invasive aneuploidy detection techniques based on cfDNA in maternal plasma and placental mRNA have made great progress over the past decade.[132] Placental miRNAs are released by exosomes and exported from the syncytiotrophoblast into the maternal circulation. A study found that 17 miRNAs were present in the placenta at concentrations at least 10-fold higher than those in maternal blood and were undetectable in maternal plasma after delivery, with miR-141, miR-149, miR-299–5p, and miR-135b being the four most abundant placental miRNAs present in maternal plasma.[133] In a case-control study, miRNAs in the plasma of PE and healthy pregnant women were examined using droplet digital PCR, and elevated levels of placenta-specific miR518b were found and could be used as a potential biomarker for PE.[134] Given that miRNAs are stable in plasma, detection of placental-derived miRNA expression can be used to assess placental and fetal development and is expected to be a biomarker. In summary, by directly targeting the analysis of fetal/placental DNA and RNA in maternal plasma (rather than the indirect analysis of placental proteins), a more definitive diagnosis can be produced, and this non-invasive assay offers the possibility to assess the occurrence of fetal-originated diseases.

Epigenetic modification changes

The placenta is a more accessible tissue than the fetus and has unique global and site-specific DNA methylation patterns. Unlike embryonic cells (inner cell mass), cells of the trophectoderm (cells that develop into the placenta) do not undergo extensive remethylation after demethylation, thus maintaining a genome-wide hypomethylated state.[135] Since placental DNA methylation may result from the hypomethylation of the early embryo, the disturbance of the specific DNA methylation pattern may reflect the abnormal programming of early embryonic development. A growing number of studies have found that detecting the DNA methylation characteristics of the placenta can serve as a biomarker to predict long-term diseases.[136139] Histone modification is involved in gene transcriptional regulation, and abnormal histone modification status is commonly implicated with pregnancy-related diseases and placental dysplasia. A genome-wide analysis of healthy human placentas and FGR placentas showed that changes in placental histone 3 lysine 27 acetylation are associated with placental function and early fetal development.[140] Therefore, epigenetic results of gene-environment interactions in the placenta may serve as biomarkers for abnormal fetal development and susceptibility to long-term diseases.

Combined screening of maternal characteristics, serum biomarkers, and fetal biometric parameters

At present, the sensitivity and specificity of clinical detection methods for FGR are not ideal. It is estimated that close to 50% of FGR cases are missed prenatally.[141] Fetal biometric parameters (ultrasound measurements) could not distinguish healthy but small fetuses from FGR fetuses. There are also no serum biomarkers that are accurate enough to be used clinically as a single predictive marker. Therefore, many researchers have proposed combined models to obtain better predictive values. The combined screening approach can reflect a variety of pregnancy conditions: maternal characteristics (such as gestational age and disease) can help identify high-risk individuals and be closely monitored, Doppler ultrasound of the uterine artery can indicate the adequacy of spiral arterial infiltration, and biomarker levels reflect impaired placental secretory function. Several studies have found that using a combination of maternal characteristics, serum biomarkers, and fetal biometrics may improve predictive correlations.[142144]

PLACENTAL BIOMARKERS OF FETAL-ORIGINATED DISEASES

Current clinical detection techniques are still suboptimal in identifying the risk of FGR and predicting long-term diseases, so there is an urgent need to explore more potential biomarkers and detection techniques for early identification of fetal dysplasia, timely improvement of pregnancy outcomes, and intervention of long-term disease susceptibility.

Placental biomarkers of fetal growth restriction

FGR is a leading cause of stillbirth and is strongly associated with increased perinatal morbidity. There is no effective intrauterine treatment, so it relies on early detection and timely delivery. Some specific factors reflecting placental and fetal development can be secreted into the maternal circulation, and these specific factors have the potential to be biomarkers for early diagnosis of FGR (Table 3).

Table 3: Potential placental biomarkers of fetal growth restriction in humans
Placental biomarkers Detection sample References
PlGF/sFlt1 Maternal plasma [145147]
PAPP-A Maternal plasma [127,148]
sEng Maternal plasma [149151]
VEGF Placenta/ Maternal plasma [152,153]
IGF2 Maternal plasma /Placenta [87,154]
IGFBP-4 Placenta [155]
ADAM12 Maternal plasma [127,148]
PGH Maternal plasma [87,156]
SPINT1 Maternal plasma [157159]
DLK1 Maternal plasma [160]
sICAM-1, sVCAM-1 Maternal plasma [161]
PP-13 Maternal plasma [128,162]
PlGF, placental growth factor; sFlt1, soluble fms-like tyrosine kinase 1; PAPP-A, pregnancy-associated plasma protein-A; sEng, soluble endoglin; VEGF, vascular endothelial growth factor; IGF2, insulin-like growth factor 2; IGFBP-4, IGF binding protein-4; ADAM12, a disintegrin and metalloprotease 12; PGH, placental growth hormone; SPINT1, serine peptidase inhibitor Kunitz type 1; DLK1, delta-like noncanonical Notch ligand 1; sICAM-1, serum soluble intercellular adhesion molecule-1; sVCAM-1, soluble vascular cell adhesion molecule-1; PP-13, placental protein.

Pregnancy-associated plasma protein-A

Pregnancy-associated plasma protein-A (PAPP-A) is a protein hydrolase produced by the placenta, and its plasma level depends on placental volume and function. In 2005, A study of 4, 390 women with single pregnancies showed that low levels of maternal PAPP-A during 11–13 weeks of gestation were significantly associated with adverse pregnancy outcomes.[163] Cohort studies have shown that low levels of PAPP-A in maternal blood are associated with neonatal birth weight, gestational age, and preterm birth and that β-hCG is an independent predictor of gestational hypertension and PE.[164,165] Notably, although PAPP-A has a certain detection sensitivity for the occurrence of FGR, its predictive power is not sufficient for screening for FGR alone. Since then, a prospective cohort study was established to evaluate a multiparametric screening model in which maternal blood PAPP-A, β-hCG, maternal mean arterial pressure, and uterine artery Doppler parameters increased the sensitivity of the detection of early-onset small for gestational age (SGA) infants to 73%. However, the detection sensitivity of late-onset SGA was low (Table 4).[166]

Table 4: Potential human placental biomarkers of fetal-originated diseases in humans
Long-term disease Placental biomarkers Detection object References
Cardiovascular system Heart development DNA methylation of HEY2, ISL1, SRF, ACTC1, HEYL Placenta [139]
Ventricular septal defect miR-191, miR-548F1, miR-148A, miR-423, miR-92B, miR-611, miR-2110, miR-548H4 Placenta [139]
Tetralogy of Fallot DNA cytosine methylation of ARHGAP22, CDK5, TRIM27, IER3 Placenta [175]
Congenital heart disease DNA methylation of TLL1, CRABP1, FDFT1, PCK2 Placenta [176]
Conical heart defect DNA methylation of HOXD9, CNN1, NOTCH1, ECE1 Placenta [177]
Neurological behavior Autism spectrum disorder DNA methylation of CYP2E1 and IRS2 Placenta [138]
Autism spectrum disorder Expression of NHIP gene Placenta [178]
Autism spectrum disorder DNA methylation changes of NOS1AP, MOSPD1, AFAP1L2, GRIPAP1 Placenta [179]
Adverse neurobehavioral outcomes DNA methylation of HSD11B2 Placenta [98,180]
Schizophrenia ZNF664, KLC1, MAN2A2, CTDSPL, MSI2 Placenta/ Maternal plasma [181]
Neurobehavioral changes at birth miR-509–3p and miR-193b-5p Placenta [182]
Neurodevelopmental disorders OGT Placenta [183,184]
Others Pulmonary development DNA methylation changes of JAK2 Placenta [185,186]
Cryptorchidism and hypospadias in neonates Antiandrogen activity or exogenous estrogen levels Placenta
HEY2, hairy and enhancer of split-related with YRPW motif 2; ISL1, LIM-homeodomain transcription factor Islet-1; SRF, serum response factor; ACTC1, cardiac alpha-actin; HEYL, hairy and enhancer of split-related with YRPW motif-Like; ARHGAP22, RhoGAP protein; CDK5, cyclin-dependent kinase 5; TRIM27, tripartite motif-containing 27; IER3, immediate early response 3; TLL1, tolloid-like 1; CRABP1, cellular retinoid-binding protein 1; FDFT1, farnesyl-diphosphate farnesyltransferase 1; PCK2, phosphoenolpyruvate carboxykinase; HOXD9, homeobox D9; CNN1, calponin 1; NOTCH1, Notch receptor 1; ECE1, endothelin converting enzyme-1; CYP2E1, cytochrome P450 2E1; IRS2, insulin receptor substrate 2; NHIP, LOC105373085; NOS1AP, nitric oxide synthase-1 adaptor protein; MOSPD1, major sperm protein domain‐containing proteins 1; AFAP1L2, actin filament associated protein 1 like 2; GRIPAP1, GRIP1 associated protein 1; HSD11B2, 11beta-hydroxysteroid dehydrogenase type 2; ZNF664, zinc finger protein 664; KLC1, kinesin light chain 1; MAN2A2, mannosidase alpha class 2A member 2; CTDSPL, CTD small phosphatase like; MSI2, musashi RNA binding protein 2; OGT, O-GlcNAc transferase; JAK2, Janus kinase 2.

Pro-angiogenic and anti-angiogenic factors

The placenta is one of the most vascularized organs. The imbalance between pro-angiogenic and anti-angiogenic factors is thought to be an important trigger for placental dysfunction and FGR. Soluble endoglin (sEng) is a placental-derived anti-angiogenic factor. A prospective case-control study showed that maternal plasma biochemical parameters (sFlt-1, sEng, and PAPP-A) combined with uterine artery Doppler parameters provide a better prediction of the degree of placental vascular injury and assess the risk of fetal FGR.[126] Longitudinal case-control studies have also demonstrated that imbalances in maternal PlGF and anti-angiogenic factors (sEng and sVEGFR-1) increase the risk of delivering SGA newborns and/or the occurrence of PE. Continuous determination of PlGF, sEng, and soluble vascular endothelial growth factor receptor 1 (sVEGFR-1) in early and second trimesters is valuable for the risk assessment of PE and SGA.[167] Vascular endothelial growth factor (VEGF) plays a vital role in placental vasculogenesis and angiogenesis. Clinical studies have shown that VEGF levels in maternal blood and placenta are positively correlated with neonatal birth weight and may have the potential to predict the occurrence of FGR.[153,168] Another study also showed that maternal peripheral blood VEGF levels exhibited the highest predictive value for increased risk of FGR in the third trimester.[152] In addition, serum soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1), which reflects vascular endothelial function, also have the potential to predict pregnancy comorbidities and adverse pregnancy outcomes. In a prospective longitudinal study, sICAM-1 and sVCAM-1 in maternal circulation during mid-gestation were highly predictive of PE or other pregnancy comorbidities in late-gestation, with predictive sensitivities of 50% and 46% for FGR and preterm labor, respectively.[161]

Insulin-like growth factor and its receptor and binding protein

IGF and its receptors and binding proteins can be expressed in the placenta and released into the maternal circulation. There is growing evidence that abnormal expression of IGF-related genes is involved in the occurrence of FGR. A prospective cohort study that collected consecutive maternal samples from a low-risk population at 28 weeks gestation and followed fetal growth until delivery found that the mRNA expression of six growth factors (including IGF1, IGF2, PGH, IGFBP2, IGF1R, ADAM12) was dysregulated in the blood of mothers carrying severely preterm fetuses with FGR. Notably, as early as 28 weeks of gestation, the mRNA that codes for growth genes in the maternal circulation was dysregulated.[87] It suggests that measurement of IGF-related genes at 28 weeks of gestation, even when an ultrasound shows normal fetal size at that time, can identify pregnancies at risk for full-term FGR and may be a novel biomarker for predicting and diagnosing FGR. IGF binding protein-4 (IGFBP-4) is involved in the regulation of IGF bioavailability. Maternal serum IGFBP-4 levels may be implicated in abnormal placental growth and fetal development and have the potential to be an early biomarker of FGR.[155] Placental growth hormone (PGH) and a disintegrin and metalloprotease 12 (ADAM12) are placental-derived growth factors that are also involved in the regulation of IGF-related signaling pathways. It was found that PGH2 mRNA levels in the maternal circulation may be related to fetal growth and are significantly correlated with fetal biometrics and birth weight.[156] However, another study suggested that maternal serum PGH at 11–13 weeks of gestation is unlikely to be a useful biochemical marker for early prediction of SGA.[169] Maternal ADAM12 and PAPP-A concentrations were reduced in SGA and all neonates weighing less than 2.5 kg, with a linear relationship between the severity of FGR and reductions in ADAM12 and PAPP-A.[148] In early pregnancy screening, the detection rate of FGR using a combination of maternal blood PAPP-A, ADAM12 levels, and uterine artery Doppler analysis was 68%.[127]

Others

There are also several bioactive factors derived from the placenta that can predict FGR, but fewer studies have been published and their application in the diagnosis of FGR remains to be explored. For example, delta-like noncanonical Notch ligand 1 (DLK1) is a cell growth regulator. Clinical and rodent studies have shown that placental and fetal-derived DLK1 in the maternal circulation may serve as a potential marker of FGR and SGA.[160,170] However, other studies have concluded to the contrary, that although circulating DLK1 levels are reduced in mid-gestation in pregnant women delivering newborns with SGA, DLK1 alone does not predict the occurrence of SGA.[171]

Serine peptidase inhibitor Kunitz type 1 (SPINT1) is a highly expressed placental protein. Prospective cohort studies have identified low-circulating SPINT1 as a marker of placental insufficiency and a novel biomarker for predicting fetal dysplasia. SPINT1 has a positive predictive value ranging from 10% to 38% for newborns below the 3rd, 5th, and 10th percentiles of birth weight.[157,158] In addition, it has been noted that SPINT2 can also be used as a biomarker to identify pregnancies with an elevated risk of stillbirth, such as PE combined with FGR, but its predictive potential for FGR is lower than that of its homolog, SPINT1.[159]

PE is a pregnancy complication involving placental impairment and/or placental dysfunction that can lead to an increased risk of FGR.[172] Placenta-derived PlGF and sFlt-1 in maternal blood have been recommended by several countries and organizations as diagnostic and/or predictive biomarkers for PE.[173] Some studies have also found that the sFlt-1 / PlGF ratio can be used both for the diagnosis of PE and to predict the occurrence of PE-induced FGR, but its predictive potential is controversial.[145147]

Placental protein 13 (PP-13) is highly expressed in the placenta and is involved in placental implantation, vascular invasion, and remodeling. Chaftez et al. found that low levels of PP-13 in early pregnancy had a detection sensitivity of 79% for PE, 33% for IUGR, and 28% for preterm birth, respectively.[128] In another study, based on a 5% fixed false positive rate, the sensitivity of maternal circulating PP-13 in early pregnancy to PE, early-onset PE, and SGA was 24%, 45%, and 26%, respectively.[162]

In addition, a large-scale retrospective study in 2020 sequenced the whole genome of maternal plasma cfDNA from 2, 199 pregnancies and found different cfDNA promoter coverage patterns between healthy and pregnancies with comorbidities (macrosomia, FGR, gestational diabetes mellitus, and PE).[174] In the construction and validation of classifiers based on a literature search, it was found that the prediction accuracy of multi-gene combinations for FGR could reach 70.0% to 79.5%.

In summary, the in-depth study of placental biomarkers is highly likely to develop early warning and prevention measures for FGR, which will help to improve adverse pregnancy outcomes and ensure normal fetal development.

Placental biomarkers for long-term diseases

Given the existence of the placenta-fetal organ axis and the synchronous disruption of placental and fetal development in response to adverse environmental exposures, placental biomarkers may have the potential to predict the occurrence of long-term diseases. Given the existence of the placenta-fetal organ axis and the synchronous disruption of placental and fetal development in response to adverse environmentalexposures, placental biomarkers may have the potential to predict the occurrence of long-term diseases (Table 4).

Cardiovascular system diseases

Currently, there are no biomarkers available in clinical practice for the prenatal or postnatal detection of congenital heart defects. Studies of the Helsinki birth cohort have found that placental phenotypic characteristics such as size, shape, and efficiency are strong predictors of cardiovascular disease in offspring.[70,110,111] It is reported that in the placenta of the fetal ventricular septal defect (VSD), there was likely to be a significant minority of cytosine loci in which parallel or correlated epigenetic modifications could be identified in cardiac development genes in general and specific genes related to ventricular development in the placenta from VSD pregnancies, and ultimately the study identified 1, 488 unique loci (one per gene).[139] These loci were significantly differentially methylated in VSD placenta, with 80 highly accurate potential cytosine-guanine (CpG) sites are available for detection of VSD (AUC= 1.0 at FDR P < 0.005), including genes involved in ventricular development (HEY2, ISL1), cardiac circulation (SRF), cardiomyocyte differentiation (ACTC1, HEY2), and cardiac septal development (ISL1), cardiac morphogenesis (SRF, HEY2, ISL1, HEYL), Notch signaling pathway (HEY2, HEYL), cardiac ventricular development (ISL1) and myocardial tissue development (ACTC1, ISL1). In addition, methylation changes in 8 miRNAs (including miR-191, miR-548F1, miR-148A, miR-423, miR-92B, miR-611, miR-2110, and miR-548H4) were identified alter cardiac development programming at the post-transcriptional level and serve as biomarkers for predicting fetal VSD. A retrospective study identified several novel placental CpG biomarkers with excellent predictive accuracy (AUC ≥ 0.95; 95% CI) for tetralogy of Fallot, where the four CpG loci with the best predictive performance were located on the ARHGAP22, CDK5, TRIM27, and IER3 genes.[175] In addition, abnormal DNA methylation levels of TLL1, CRABP1, FDFT1, and PCK2 in placental tissue are also closely related to fetal congenital heart disease.[176] Another clinical study also found that altered DNA methylation levels of HOXD9, CNN1, NOTCH1, and ECE1 in the placenta are associated with conotruncal heart defects and may be used as potential epigenetic biomarkers to detect conotruncal heart defects.[177]

Nervous system diseases

The brain is one of the organs most susceptible to the effects of the placenta, and the origin of multiple neurobehavioral disorders may be attributed to pathological changes in the placenta.[190,191] Placental lesions (such as increased syncytial knots, and chorioamnionic edema) as predictors of cerebral palsy and neurocognitive abnormalities in very low birth weight school-age children.[192] A prospective cohort study found that DNA methylation levels of placental cytochrome P450 2E1 (CYP2E1) and insulin receptor substrate 2 (IRS2) may serve as early warning indicators of autism in children.[138] Another study that conducted a full methylomics analysis of placentas in the prospective MARBLES cohort found that methylation levels of placental CYP2E1 and IRS2 DMR were genetic and environmental modifiers associated with a high risk of autistic spectrum disorder (ASD), which is expected to serve as a biomarker for early intervention and prevention of ASD.[138] In addition, a novel gene, LOC105373085 (renamed NHIP due to neuronal hypoxia induction), present within chromosome 22q13.33, is expressed in both the placenta and neurons in response to hypoxia and may be a new gene regulatory locus for ASD.[178] Another genome-wide methylation analysis of full-term placental tissue for ASD subtypes identified five placental markers with high accuracy for ASD: NOS1AP, MOSPD1, AFAP1L2, and GRIPAP1.[179]

A transcriptome-wide association study of healthy full-term placentas (N= 147) screened for multiple candidate placental genes associated with offspring schizophrenia: ZNF664 (preferred), KLC1, MAN2A2, CTDSPL, and MSI2, which can be detected in maternal blood (cfRNA) and can be used to predict pregnancy outcomes or offspring schizophrenia.[181] In addition, placental 11β-hydroxysteroid dehydrogenase 2 (11β-HSD2) expression was affected by the intrauterine environment, and its DNA methylation was correlated with neonatal neurobehavior. Assessing the degree of HSD11B2 (11β-hydroxysteroid dehydrogenase 2 coding gene) methylation in the placenta can prospectively predict psychological, behavioral, and cognitive problems in later childhood.[98,180] miRNAs also have the potential to predict neurological disorders in offspring. A study of full-term placentas found that increased placental miR-16 expression was negatively associated with neonatal attention scores and miR-146a and miR-182 were positively correlated with neonatal motor quality scores.[193] Another observational study also showed that altered placental miR-509–3p and miR-193b-5p abundance associated with cadmium exposure may be predictive of atypical neurobehavioral outcomes in newborns.[182]

Others

The main determinant of intrauterine bone mineralization is fetal blood calcium concentration, which is dependent on placental calcium ion transport. A cohort study suggests that IUGR placenta size and impaired mineral supply are associated with reduced bone size after birth and an increased risk of long-term fracture.[122] It has also been shown that the mRNA expression level of calcium transporter proteins in the placental basal membrane is positively correlated with the whole-body bone mineral content of offspring.[194] Although the mechanism is unknown, it can be evaluated as an independent risk factor for poor fetal bone growth, short stature in childhood, and increased susceptibility to osteoporosis in adulthood, and provides new ideas for the study of placental biomarkers.

Boys with low placental weight at birth have a higher risk of cryptorchidism and hypospadias, according to a Danish cohort study.[187] A case-control study found that antiandrogen biological activity in the placenta at delivery was significantly associated with the risk of neonatal cryptorchidism/hypospadias in males and may be a biomarker for genitourinary malformation.[188] In addition, a prospective maternal and infant cohort study also suggests that the concentration of exogenous estrogen accumulated in the placenta may play a role in assessing and predicting the risk of cryptorchidism and/or hypospadias.[189]

In summary, although the development process of the placenta is not entirely consistent with that of the fetus, the molecular characteristics of the placenta may reveal the intrauterine development and long-term health outcomes of the fetus to a certain extent. Although the specific mechanisms by which the placenta-fetal organ axis exerts its influence are unknown, numerous current studies have provided strong evidence for a link between placental response and offspring health/disease status and may serve as biomarkers to identify which children are at risk of fetal-originated diseases.

SUMMARY AND PROSPECT

In conclusion, adverse environments can lead to fetal dysplasia and susceptibility to a variety of fetal-originated diseases by affecting placental development or synchronously disrupting placental-fetal development. Biomarkers that can reflect placental dysfunction can be used for non-invasive detection of fetal organ development and have the potential to predict susceptibility to fetal-originated diseases (Figure 1). However, the mechanisms underlying the susceptibility to fetal-originated diseases due to abnormal placental development are not well understood, and the targets that have been established all have significant limitations. Before these new findings can be translated into clinically relevant technologies, more basic research is needed to further explore the pathogenesis and key targets. An increasing number of "candidate" biomarkers will be identified as a result of continued research into the mechanism of placenta origin of fetal-originated diseases and the ongoing development of related experimental technologies. This will pave the way for the early diagnosis, multi-organ development assessment of offspring, early warning, and comprehensive prevention and treatment of fetal-originated diseases.

Figure 1

Figure 1. Placental biomarkers have the potential to predict susceptibility to fetal-originated diseases.

Declaration

Author contributions

All authors were involved in preparing the manuscript with contributorship: Conception and design PY, SC, HW; Manuscript draft PY, SC; Critical revision of manuscript PY, HW.

Ethics approval

Not applicable.

Source of funding

This work was supported by the National Key R&D Program of China (No 2020YFA0803900), and the National Natural Science Foundation of China (No. 82030111, 81673524)(to HW).

Conflict of interest

The authors in preparing this manuscript did not receive any compensation from any source and declare that they have no potential conflict of interest. None of the authors have been involved in legal or regulatory matters related to the paper's contents. All authors approved the final manuscript draft and are fully responsible for the writing and content of the manuscript.

Data availability statement

All data generated or analyzed during this study are included in this published article.

REFERENCES

  1. Uauy R, Kain J, Corvalan C. How can the Developmental Origins of Health and Disease (DOHaD) hypothesis contribute to improving health in developing countries? Am J Clin Nutr 2011;94:1759.
  2. Hedderson MM, Ferrara A, Avalos LA, et al. The Kaiser Permanente Northern California research program on genes, environment, and health (RPGEH) pregnancy cohort:study design, methodology and baseline characteristics. BMC Pregnancy Childbirth 2016;16:381.
  3. Fleming TP, Sun C, Denisenko O, et al. Environmental Exposures around Conception:Developmental Pathways Leading to Lifetime Disease Risk. Int J Environ Res Public Health 2021;18:9380.
  4. Drwal E, Rak A, Gregoraszczuk EL. Review:Polycyclic aromatic hydrocarbons (PAHs)-Action on placental function and health risks in future life of newborns. Toxicology 2019;411:133–142.
  5. Morales-Rubio RA, Alvarado-Cruz I, Manzano-León N, et al. In utero exposure to ultrafine particles promotes placental stress-induced programming of renin-angiotensin system-related elements in the offspring results in altered blood pressure in adult mice. Part Fibre Toxicol 2019;16:7.
  6. Ghosh R, Causey K, Burkart K, Wozniak S, Cohen A, Brauer M. Ambient and household PM2.5 pollution and adverse perinatal outcomes:A meta-regression and analysis of attributable global burden for 204 countries and territories. PLoS Med 2021;18:e1003718.
  7. Arias A, Schander JA, Bariani MV, et al. Dexamethasone-induced intrauterine growth restriction modulates expression of placental vascular growth factors and fetal and placental growth. Mol Hum Reprod 2021;27:gaab006.
  8. Bar-Lev MR, Maayan-Metzger A, Matok I, Heyman Z, Sivan E, Kuint J. Short-term outcomes in low birth weight infants following antenatal exposure to betamethasone versus dexamethasone. Obstet Gynecol 2004;104:484–488.
  9. Xiong YW, Zhu HL, Nan Y, et al. Maternal cadmium exposure during late pregnancy causes fetal growth restriction via inhibiting placental progesterone synthesis. Ecotoxicol Environ Saf 2020;187:109879.
  10. Zhu HL, Shi XT, Xu XF, et al. Environmental cadmium exposure induces fetal growth restriction via triggering PERK-regulated mitophagy in placental trophoblasts. Environ Int 2021;147:106319.
  11. Liu L, Liu F, Kou H, et al. Prenatal nicotine exposure induced a hypothalamic-pituitary-adrenal axis-associated neuroendocrine metabolic programmed alteration in intrauterine growth retardation offspring rats. Toxicol Lett 2012;214:307–313.
  12. Feng JH, Yan YE, Liang G, et al. Maternal and fetal metabonomic alterations in prenatal nicotine exposure-induced rat intrauterine growth retardation. Mol Cell Endocrinol 2014;394(1–2):59–69. DOI:10.1016/j.mce.2014.06.016]    DOI: 10.1016/j.mce.2014.06.016
  13. Connor S, Tan KY, Pestell CF, Fitzpatrick JP. The Demographic and Neurocognitive Profile of Clients Diagnosed With Fetal Alcohol Spectrum Disorder in PATCHES Paediatrics Clinics Across Western Australia and the Northern Territory. Alcohol Clin Exp Res 2020;44:1284–1291.
  14. He Z, Zhang J, Chen G, et al. H19/let-7 axis mediates caffeine exposure during pregnancy induced adrenal dysfunction and its multi-generation inheritance. Sci Total Environ 2021;792:148440.
  15. Jafari A, Naghshi S, Shahinfar H, et al. Relationship between maternal caffeine and coffee intake and pregnancy loss:A grading of recommendations assessment, development, and evaluation-assessed, dose-response meta-analysis of observational studies. Front Nutr 2022;9:886224.
  16. Janardhan N, Roy PG, Vyas AK. Impact of maternal substance use on offspring's cardiovascular health. Toxicol Appl Pharmacol 2022;450:116164.
  17. Cestonaro C, Menozzi L, Terranova C. Infants of Mothers with Cocaine Use:Review of Clinical and Medico-Legal Aspects. Children (Basel) 2022;9(1):67.    DOI: 10.3390/children9010067    PMID: 35053692
  18. Velez ML, Jordan C, Jansson LM. Reconceptualizing non-pharmacologic approaches to Neonatal Abstinence Syndrome (NAS) and Neonatal Opioid Withdrawal Syndrome (NOWS):A theoretical and evidence-based approach. Part II:The clinical application of nonpharmacologic care for NAS/NOWS. Neurotoxicol Teratol 2021;88:107032.
  19. Albano GD, La Spina C, Pitingaro W, et al. Intrauterine and Neonatal Exposure to Opioids:Toxicological, Clinical, and Medico-Legal Issues. Toxics 2023;11:62.
  20. Hawkins SJ, Crompton LA, Sood A, et al. Nanoparticle-induced neuronal toxicity across placental barriers is mediated by autophagy and dependent on astrocytes. Nat Nanotechnol 2018;13:427–433.
  21. Choi H, Rauh V, Garfinkel R, Tu Y, Perera FP. Prenatal exposure to airborne polycyclic aromatic hydrocarbons and risk of intrauterine growth restriction. Environ Health Perspect 2008;116:658–665.
  22. Perera FP, Rauh V, Tsai WY, et al. Effects of transplacental exposure to environmental pollutants on birth outcomes in a multiethnic population. Environ Health Perspect 2003;111:201–205.
  23. Li R, Wang X, Wang B, et al. Gestational 1-nitropyrene exposure causes fetal growth restriction through disturbing placental vascularity and proliferation. Chemosphere 2018;213:252–258.
  24. Yingya Zhao, Qi Song, Wenzhen Ge, et al. Associations between in utero exposure to polybrominated diphenyl ethers, pathophysiological state of fetal growth and placental DNA methylation changes. Environ Int 2019;133:105255.
  25. Jin Y, Li J, Deng X, et al. Association between fetal growth restriction and maternal exposure to polybrominated diphenyl ethers. Ecotoxicol Environ Saf 2020;198:110623.
  26. Yu Z, Han Y, Shen R, et al. Gestational di-(2-ethylhexyl) phthalate exposure causes fetal intrauterine growth restriction through disturbing placental thyroid hormone receptor signaling. Toxicol Lett 2018;294:1–10.
  27. Guo C, Yang Y, Shi MX, et al. Critical time window of fenvalerate-induced fetal intrauterine growth restriction in mice. Ecotoxicol Environ Saf 2019;172:186–193.
  28. Wang B, Liu JJ, Wang Y, et al. Maternal Fenvalerate Exposure Induces Fetal Intrauterine Growth Restriction Through Disrupting Placental Thyroid Hormone Receptor Signaling. Toxicol Sci 2017;157:377–386.
  29. Sabra S, Malmqvist E, Saborit A, Gratacos E, Gomez Roig MD. Heavy metals exposure levels and their correlation with different clinical forms of fetal growth restriction. PLoS One 2017;12:e0185645.
  30. Camsari C, Folger JK, McGee D, et al. Effects of Periconception Cadmium and Mercury Co-Administration to Mice on Indices of Chronic Diseases in Male Offspring at Maturity. Environ Health Perspect 2017;125:643–650.
  31. Müller JE, Meyer N, Santamaria CG, et al. Bisphenol A exposure during early pregnancy impairs uterine spiral artery remodeling and provokes intrauterine growth restriction in mice. Sci Rep 2018;8:9196.
  32. Nava-Castro KE, Ramírez-Nieto R, Méndez-García LA, et al. Environmental Pollution as a Risk Factor in Testicular Tumour Development:Focus on the Interaction between Bisphenol A and the Associated Immune Response. Int J Environ Res Public Health 2019;16:4113.
  33. Levario-Carrillo M, Amato D, Ostrosky-Wegman P, González-Horta C, Corona Y, Sanin LH. Relation between pesticide exposure and intrauterine growth retardation. Chemosphere 2004;55:1421–1427.
  34. Zhao Y, Chen L, Li LX, et al. Gender-specific relationship between prenatal exposure to phthalates and intrauterine growth restriction. Pediatr Res 2014;76:401–408.
  35. Milnerowicz-Nabzdyk E, Bizon A. How does tobacco smoke influence the morphometry of the fetus and the umbilical cord?-Research on pregnant women with intrauterine growth restriction exposed to tobacco smoke. Reprod Toxicol 2015;58:79–84.
  36. Onoda A, Takeda K, Umezawa M. Pretreatment with N-acetyl cysteine suppresses chronic reactive astrogliosis following maternal nanoparticle exposure during gestational period. Nanotoxicology 2017;11:1012–1025.
  37. Carter RC, Wainwright H, Molteno CD, et al. Alcohol, Methamphetamine, and Marijuana Exposure Have Distinct Effects on the Human Placenta. Alcohol Clin Exp Res 2016;40:753–764.
  38. Singh M, Varukolu S, Chauhan A, et al. Paracetamol exposure and asthma:What does the evidence say? An overview of systematic reviews. Pediatr Pulmonol 2021;56:3189–3199.
  39. Ryan L, Ehrlich S, Finnegan L. Cocaine abuse in pregnancy:effects on the fetus and newborn. Neurotoxicol Teratol 1987;9:295–299.
  40. Xiao D, Huang X, Xu Z, Yang S, Zhang L. Prenatal cocaine exposure differentially causes vascular dysfunction in adult offspring. Hypertension 2009;53:937–943.
  41. Pacheco ALD, de Melo IS, de Souza FMA, et al. Maternal crack cocaine use in rats leads to depressive- and anxiety-like behavior, memory impairment, and increased seizure susceptibility in the offspring. Eur Neuropsychopharmacol 2021;44:34–50.
  42. de Castro A, Jones HE, Johnson RE, Gray TR, Shakleya DM, Huestis MA. Maternal methadone dose, placental methadone concentrations, and neonatal outcomes. Clin Chem 2011;57:449–458.
  43. Hulse GK, Milne E, English DR, Holman CD. The relationship between maternal use of heroin and methadone and infant birth weight. Addiction 1997;92(11):1571–1579.
  44. Fishman B, Daniel S, Koren G, Lunenfeld E, Levy A. Pregnancy outcome following opioid exposure:A cohort study. PLoS One 2019;14:e0219061.
  45. Straub HL, Mou J, Drennan KJ, Pflugeisen BM. Maternal Marijuana Exposure and Birth Weight:An Observational Study Surrounding Recreational Marijuana Legalization. Am J Perinatol 2021;38:65–75.
  46. Smid MC, Metz TD, McMillin GA, et al. Prenatal Nicotine or Cannabis Exposure and Offspring Neurobehavioral Outcomes. Obstet Gynecol 2022;139:21–30.
  47. Paul SE, Hatoum AS, Fine JD, et al. Associations Between Prenatal Cannabis Exposure and Childhood Outcomes:Results From the ABCD Study. JAMA Psychiatry 2021;78:64–76.
  48. Doi M, Nakama N, Sumi T, Usui N, Shimada S. Prenatal methamphetamine exposure causes dysfunction in glucose metabolism and low birthweight. Front Endocrinol (Lausanne) 2022;13:1023984.
  49. Dong N, Zhu J, Han W, et al. Maternal methamphetamine exposure causes cognitive impairment and alteration of neurodevelopment-related genes in adult offspring mice. Neuropharmacology 2018;140:25–34.
  50. Sanjari Moghaddam H, Mobarak Abadi M, Dolatshahi M, et al. Effects of Prenatal Methamphetamine Exposure on the Developing Human Brain:A Systematic Review of Neuroimaging Studies. ACS Chem Neurosci 2021;12:2729–2748.
  51. Lu X, Mao T, Dai Y, et al. Azithromycin exposure during pregnancy disturbs the fetal development and its characteristic of multi-organ toxicity. Life Sci 2023;329:121985.    DOI: 10.1016/j.lfs.2023.121985    PMID: 37516432
  52. Flory T Muanda, Odile Sheehy, Anick Bérard. Use of antibiotics during pregnancy and risk of spontaneous abortion. CMAJ 2017;189:E625–E633.
  53. Voormolen DN, de Wit L, van Rijn BB, et al. Neonatal Hypoglycemia Following Diet-Controlled and Insulin-Treated Gestational Diabetes Mellitus. Diabetes Care 2018;41:1385–1390.
  54. Fraser A, Nelson SM, Macdonald-Wallis C, Sattar N, Lawlor DA. Hypertensive disorders of pregnancy and cardiometabolic health in adolescent offspring. Hypertension 2013;62:614–620.
  55. Zheng W, Zhang X, Feng Y, et al. Association of Corpus Callosum Development With Fetal Growth Restriction and Maternal Preeclampsia or Gestational Hypertension. JAMA Netw Open 2022;5:e2226696.
  56. Ayano G, Betts K, Tait R, Dachew BA, Alati R. Maternal depressive and anxiety symptoms and the risk of attention deficit hyperactivity disorder symptoms in offspring aged 17:Findings from the Raine Study. J Affect Disord 2021;284:149–156.
  57. Zhu Q, Yang X, Zhang Y, Shan C, Shi Z. Role of the Gut Microbiota in the Increased Infant Body Mass Index Induced by Gestational Diabetes Mellitus. mSystems 2022;7:e0046522.
  58. Chu AHY, Godfrey KM. Gestational Diabetes Mellitus and Developmental Programming. Ann Nutr Metab 2020;76 Suppl 3:4-15.
  59. Furse S, Koulman A, Ozanne SE, Poston L, White SL, Meek CL. Altered Lipid Metabolism in Obese Women With Gestational Diabetes and Associations With Offspring Adiposity. J Clin Endocrinol Metab 2022;107:e2825-e2832.
  60. Kim C, Harrall KK, Glueck DH, Needham BL, Dabelea D. Gestational diabetes mellitus, epigenetic age and offspring metabolism. Diabet Med 2022;39:e14925.
  61. Yu Y, Arah OA, Liew Z, et al. Maternal diabetes during pregnancy and early onset of cardiovascular disease in offspring:population based cohort study with 40 years of follow-up. BMJ 2019;367:l6398.
  62. Yamamoto JM, Benham JL, Dewey D, et al. Neurocognitive and behavioural outcomes in offspring exposed to maternal pre-existing diabetes:a systematic review and meta-analysis. Diabetologia 2019;62:1561-1574.
  63. Smith CA. Effects of maternal under nutrition upon the newborn infant in Holland (1944-1945). J Pediatr 1947;30:229-243.
  64. Sarli PM, Manousopoulou A, Efthymiou E, et al. Liver Proteome Profile of Growth Restricted and Appropriately Grown Newborn Wistar Rats Associated With Maternal Undernutrition. Front Endocrinol (Lausanne) 2021;12:684220.
  65. Zouridis A, Manousopoulou A, Potiris A, et al. Impact of Maternal Food Restriction on Heart Proteome in Appropriately Grown and Growth-Restricted Wistar-Rat Offspring. Nutrients 2021;13:466.
  66. Potiris A, Manousopoulou A, Zouridis A, et al. The Effect of Prenatal Food Restriction on Brain Proteome in Appropriately Grown and Growth Restricted Male Wistar Rats. Front Neurosci 2021;15:665354.
  67. Hagstrom H, Simon TG, Roelstraete B, Stephansson O, Soderling J, Ludvigsson JF. Maternal obesity increases the risk and severity of NAFLD in offspring. J Hepatol 2021;75:1042-1048.
  68. Liu X, Li X, Xia B, et al. High-fiber diet mitigates maternal obesity-induced cognitive and social dysfunction in the offspring via gut-brain axis. Cell Metab 2021;33:923-938 e926.
  69. Davenport BN, Wilson RL, Jones HN. Interventions for placental insufficiency and fetal growth restriction. Placenta 2022;125:4-9.
  70. Eriksson JG, Kajantie E, Thornburg KL, Osmond C, Barker DJ. Mother's body size and placental size predict coronary heart disease in men. Eur Heart J 2011;32:2297-2303.
  71. Hu C, Wu Z, Huang Z, Hao X, Wang S, Deng J. et al. Nox2 impairs VEGF-A-induced angiogenesis in placenta via mitochondrial ROS-STAT3 pathway. Redox Biol 2021;45:102051.
  72. McIntyre KR, Hayward CE, Sibley CP, Greenwood SL, Dilworth MR. Evidence of adaptation of maternofetal transport of glutamine relative to placental size in normal mice, and in those with fetal growth restriction. J Physiol 2019;597:4975-4990.
  73. Janzen C, Lei MY, Cho J, Sullivan P, Shin BC, Devaskar SU. Placental glucose transporter 3 (GLUT3) is up-regulated in human pregnancies complicated by late-onset intrauterine growth restriction. Placenta 2013;34:1072-1078.
  74. Xu P, Wu Z, Xi Y, Wang L. Epigenetic regulation of placental glucose transporters mediates maternal cadmium-induced fetal growth restriction. Toxicology 2016;372:34-41.
  75. Jansson T, Castillo-Castrejon M, Gupta MB, Powell TL, Rosario FJ. Down-regulation of placental Cdc42 and Rac1 links mTORC2 inhibition to decreased trophoblast amino acid transport in human intrauterine growth restriction. Clin Sci (Lond) 2020;134:53-70.
  76. Chen YY, Rosario FJ, Shehab MA, Powell TL, Gupta MB, Jansson T. Increased ubiquitination and reduced plasma membrane trafficking of placental amino acid transporter SNAT-2 in human IUGR. Clin Sci (Lond) 2015;129:1131-1141.
  77. Paolini CL, Marconi AM, Ronzoni S, et al. Placental transport of leucine, phenylalanine, glycine, and proline in intrauterine growth-restricted pregnancies. J Clin Endocrinol Metab 2001;86:5427-5432.
  78. Johansson M, Karlsson L, Wennergren M, Jansson T, Powell TL. Activity and protein expression of Na+/K+ ATPase are reduced in microvillous syncytiotrophoblast plasma membranes isolated from pregnancies complicated by intrauterine growth restriction. J Clin Endocrinol Metab 2003;88:2831-2837.
  79. Chassen SS, Ferchaud-Roucher V, Gupta MB, Jansson T, Powell TL. Alterations in placental long chain polyunsaturated fatty acid metabolism in human intrauterine growth restriction. Clin Sci (Lond) 2018;132:595-607.
  80. Assumpcao RP, Mucci DB, Fonseca FCP, et al. Fatty acid profile of maternal and fetal erythrocytes and placental expression of fatty acid transport proteins in normal and intrauterine growth restriction pregnancies. Prostaglandins Leukot Essent Fatty Acids 2017;125:24-31.
  81. Loubiere LS, Vasilopoulou E, Bulmer JN, et al. Expression of thyroid hormone transporters in the human placenta and changes associated with intrauterine growth restriction. Placenta 2010;31:295-304.
  82. Chan SY, Franklyn JA, Pemberton HN, et al. Monocarboxylate transporter 8 expression in the human placenta:the effects of severe intrauterine growth restriction. J Endocrinol 2006;189:465-471.
  83. Vasilopoulou E, Loubiere LS, Martin-Santos A, et al. Differential triiodothyronine responsiveness and transport by human cytotrophoblasts from normal and growth-restricted pregnancies. J Clin Endocrinol Metab 2010;95:4762-4770.
  84. Settle P, Sibley CP, Doughty IM, et al. Placental lactate transporter activity and expression in intrauterine growth restriction. J Soc Gynecol Investig 2006;13:357-363.
  85. Harrison DJ, Creeth HDJ, Tyson HR, et al. Placental endocrine insufficiency programs anxiety, deficits in cognition and atypical social behaviour in offspring. Hum Mol Genet 2021;30:1863-1880.
  86. Whitehead CL, McNamara H, Walker SP, et al. Identifying late-onset fetal growth restriction by measuring circulating placental RNA in the maternal blood at 28 weeks' gestation. Am J Obstet Gynecol 2016;214:521 e521-521 e528.
  87. Whitehead CL, Walker SP, Mendis S, Lappas M, Tong S. Quantifying mRNA coding growth genes in the maternal circulation to detect fetal growth restriction. Am J Obstet Gynecol 2013;209:133 e131-139.
  88. Vacher CM, Lacaille H, O'Reilly JJ, et al. Placental endocrine function shapes cerebellar development and social behavior. Nat Neurosci 2021;24:1392-1401.
  89. Sato K. Placenta-derived hypo-serotonin situations in the developing forebrain cause autism. Med Hypotheses 2013;80:368-372.
  90. Ranzil S, Ellery S, Walker DW, et al. Disrupted placental serotonin synthetic pathway and increased placental serotonin:Potential implications in the pathogenesis of human fetal growth restriction. Placenta 2019;84:74-83.
  91. Li D, Gao R, Qin L, Yue H, Sang N. New Insights into Prenatal NO2 Exposure and Behavioral Abnormalities in Male Offspring:Disturbed Serotonin Metabolism and Delayed Oligodendrocyte Development. Environ Sci Technol 2022;56:11536-11546.
  92. Noorlander CW, Ververs FF, Nikkels PG, van Echteld CJ, Visser GH, Smidt MP. Modulation of serotonin transporter function during fetal development causes dilated heart cardiomyopathy and lifelong behavioral abnormalities. PLoS One 2008;3:e2782.
  93. Wu YM, Luo HW, Kou H, et al. Prenatal caffeine exposure induced a lower level of fetal blood leptin mainly via placental mechanism. Toxicol Appl Pharmacol 2015;289:109-116.
  94. Allbrand M, Eklund D, Cao Y, Nilsson K, Lodefalk M. Gene expression of leptin, leptin receptor isoforms and inflammatory cytokines in placentas of obese women- Associations to birth weight and fetal sex. Placenta 2022;117:64-71.
  95. Visan V, Balan RA, Costea CF, et al. Morphological and histopathological changes in placentas of pregnancies with intrauterine growth restriction. Rom J Morphol Embryol 2020;61:477-483.
  96. Wang T, Chen M, Yan YE, Xiao FQ, Pan XL, Wang H. Growth retardation of fetal rats exposed to nicotine in utero:possible involvement of CYP1A1, CYP2E1, and P-glycoprotein. Environ Toxicol 2009;24:33-42.
  97. Ge C, Xu D, Yu P, et al. P-gp expression inhibition mediates placental glucocorticoid barrier opening and fetal weight loss. BMC Med 2021;19:311.
  98. Marsit CJ, Maccani MA, Padbury JF, Lester BM. Placental 11-beta hydroxysteroid dehydrogenase methylation is associated with newborn growth and a measure of neurobehavioral outcome. PLoS One 2012;7:e33794.
  99. Saegusa H, Nakagawa Y, Liu YJ, Ohzeki T. Influence of placental 11beta-hydroxysteroid dehydrogenase (11beta-HSD) inhibition on glucose metabolism and 11beta-HSD regulation in adult offspring of rats. Metabolism 1999;48:1584-1588.
  100. Welberg LA, Seckl JR, Holmes MC. Inhibition of 11beta-hydroxysteroid dehydrogenase, the foeto-placental barrier to maternal glucocorticoids, permanently programs amygdala GR mRNA expression and anxiety-like behaviour in the offspring. Eur J Neurosci 2000;12:1047-1054.
  101. Kratimenos P, Penn AA. Placental programming of neuropsychiatric disease. Pediatr Res 2019;86:157-164.
  102. Behura SK, Kelleher AM, Spencer TE. Evidence for functional interactions between the placenta and brain in pregnant mice. FASEB J 2019;33:4261-4272.
  103. Redline RW. Severe fetal placental vascular lesions in term infants with neurologic impairment. Am J Obstet Gynecol 2005;192:452-457.
  104. Redline RW, O'Riordan MA. Placental lesions associated with cerebral palsy and neurologic impairment following term birth. Arch Pathol Lab Med 2000;124:1785-1791.
  105. Whitehead E, Dodds L, Joseph KS, et al. Relation of pregnancy and neonatal factors to subsequent development of childhood epilepsy:a population-based cohort study. Pediatrics 2006;117:1298-1306.
  106. Ruiz-Palacios M, Prieto-Sanchez MT, Ruiz-Alcaraz AJ, et al. Insulin Treatment May Alter Fatty Acid Carriers in Placentas from Gestational Diabetes Subjects. Int J Mol Sci 2017;18:1203.
  107. Xu P, Guo H, Wang H, et al. Downregulations of placental fatty acid transporters during cadmium-induced fetal growth restriction. Toxicology 2019;423:112-122.
  108. Goeden N, Velasquez J, Arnold KA, et al. Maternal Inflammation Disrupts Fetal Neurodevelopment via Increased Placental Output of Serotonin to the Fetal Brain. J Neurosci 2016;36:6041-6049.
  109. Camm EJ, Botting KJ, Sferruzzi-Perri AN. Near to One's Heart:The Intimate Relationship Between the Placenta and Fetal Heart. Front Physiol 2018;9:629.
  110. Barker DJ, Thornburg KL, Osmond C, Kajantie E, Eriksson JG. The surface area of the placenta and hypertension in the offspring in later life. Int J Dev Biol 2010;54:525-530.
  111. Barker DJ, Gelow J, Thornburg K, Osmond C, Kajantie E, Eriksson JG. The early origins of chronic heart failure:impaired placental growth and initiation of insulin resistance in childhood. Eur J Heart Fail 2010;12:819-825.
  112. Martyn CN, Barker DJ, Osmond C. Mothers' pelvic size, fetal growth, and death from stroke and coronary heart disease in men in the UK. Lancet 1996;348:1264-1268.
  113. Burton GJ, Jauniaux E. Pathophysiology of placental-derived fetal growth restriction. Am J Obstet Gynecol 2018;218:S745-S761.
  114. Linask KK, Han M, Bravo-Valenzuela NJ. Changes in vitelline and utero-placental hemodynamics:implications for cardiovascular development. Front Physiol 2014;5:390.
  115. Maslen CL. Recent Advances in Placenta-Heart Interactions. Front Physiol 2018;9:735.
  116. Linask KK. The heart-placenta axis in the first month of pregnancy:induction and prevention of cardiovascular birth defects. J Pregnancy 2013;2013:320413.
  117. Kaufmann P, Mayhew TM, Charnock-Jones DS. Aspects of human fetoplacental vasculogenesis and angiogenesis. II. Changes during normal pregnancy. Placenta 2004;25:114-126.
  118. Courtney JA, Cnota JF, Jones HN. The Role of Abnormal Placentation in Congenital Heart Disease;Cause, Correlate, or Consequence? Front Physiol 2018;9:1045.
  119. Wilson RL, Yuan V, Courtney JA, Tipler A, Cnota JF, Jones HN. Analysis of commonly expressed genes between first trimester fetal heart and placenta cell types in the context of congenital heart disease. Sci Rep 2022;12:10756.
  120. Woolford SJ, Curtis EM, D'Angelo S, et al. Placental volume at 11 weeks is associated with offspring bone mass at birth and in later childhood:Findings from the Southampton Women's Survey. Placenta 2020;99:101-107.
  121. Holroyd CR, Osmond C, Barker DJ, et al. Placental Size Is Associated Differentially With Postnatal Bone Size and Density. J Bone Miner Res 2016;31:1855-1864.
  122. Holroyd CR, Harvey NC, Crozier SR, et al. Placental size at 19 weeks predicts offspring bone mass at birth:findings from the Southampton Women's Survey. Placenta 2012;33:623-629.
  123. Li SH, Yin HB, Ren MR, et al. TRPV5 and TRPV6 are expressed in placenta and bone tissues during pregnancy in mice. Biotech Histochem 2019;94:244-251.
  124. Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS 2010;5:463-466.
  125. Napso T, Zhao X, Lligona MI, et al. Placental secretome characterization identifies candidates for pregnancy complications. Commun Biol 2021;4:701.
  126. Zamarian AC, Araujo Junior E, Daher S, Rolo LC, Moron AF, Nardozza LM. Evaluation of biochemical markers combined with uterine artery Doppler parameters in fetuses with growth restriction:a case-control study. Arch Gynecol Obstet 2016;294:715-723.
  127. Yu N, Cui H, Chen X, Chang Y. First trimester maternal serum analytes and second trimester uterine artery Doppler in the prediction of preeclampsia and fetal growth restriction. Taiwan J Obstet Gynecol 2017;56:358-361.
  128. Chafetz I, Kuhnreich I, Sammar M, et al. First-trimester placental protein 13 screening for preeclampsia and intrauterine growth restriction. Am J Obstet Gynecol 2007;197:35 e31-37.
  129. Lo YM, Corbetta N, Chamberlain PF, et al. Presence of fetal DNA in maternal plasma and serum. Lancet 1997;350:485-487.
  130. Poon LL, Leung TN, Lau TK, Lo YM. Presence of fetal RNA in maternal plasma. Clin Chem 2000;46:1832-1834.
  131. Tsui NB, Chim SS, Chiu RW, et al. Systematic micro-array based identification of placental mRNA in maternal plasma:towards non-invasive prenatal gene expression profiling. J Med Genet 2004;41:461-467.
  132. Go AT, van Vugt JM, Oudejans CB. Non-invasive aneuploidy detection using free fetal DNA and RNA in maternal plasma:recent progress and future possibilities. Hum Reprod Update 2011;17:372-382.
  133. Chim SS, Shing TK, Hung EC, et al. Detection and characterization of placental microRNAs in maternal plasma. Clin Chem 2008;54:482-490.
  134. Jelena M, Sopic M, Joksic I, et al. Placenta-specific plasma miR518b is a potential biomarker for preeclampsia. Clin Biochem 2020;79:28-33.
  135. Vlahos A, Mansell T, Saffery R, Novakovic B. Human placental methylome in the interplay of adverse placental health, environmental exposure, and pregnancy outcome. PLoS Genet 2019;15:e1008236.
  136. Ravaei A, Emanuele M, Nazzaro G, Fadiga L, Rubini M. Placental DNA methylation profile as predicting marker for autism spectrum disorder (ASD). Mol Med 2023;29:8.
  137. Schroeder DI, Schmidt RJ, Crary-Dooley FK, et al. Placental methylome analysis from a prospective autism study. Mol Autism 2016;7:51.
  138. Zhu Y, Mordaunt CE, Yasui DH, et al. Placental DNA methylation levels at CYP2E1 and IRS2 are associated with child outcome in a prospective autism study. Hum Mol Genet 2019;28:2659-2674.
  139. Radhakrishna U, Albayrak S, Zafra R, et al. Placental epigenetics for evaluation of fetal congenital heart defects:Ventricular Septal Defect (VSD). PLoS One 2019;14:e0200229.
  140. Paauw ND, Lely AT, Joles JA, et al. H3K27 acetylation and gene expression analysis reveals differences in placental chromatin activity in fetal growth restriction. Clin Epigenetics 2018;10:85.
  141. Pedroso MA, Palmer KR, Hodges RJ, Costa FDS, Rolnik DL. Uterine Artery Doppler in Screening for Preeclampsia and Fetal Growth Restriction. Rev Bras Ginecol Obstet 2018;40:287-293.
  142. Bakalis S, Peeva G, Gonzalez R, Poon LC, Nicolaides KH. Prediction of small-for-gestational-age neonates:screening by biophysical and biochemical markers at 30-34 weeks. Ultrasound Obstet Gynecol 2015;46:446-451.
  143. Dieste-Perez P, Saviron-Cornudella R, Tajada-Duaso M, et al. Personalized Model to Predict Small for Gestational Age at Delivery Using Fetal Biometrics, Maternal Characteristics, and Pregnancy Biomarkers:A Retrospective Cohort Study of Births Assisted at a Spanish Hospital. J Pers Med 2022;12:762.
  144. Kienast C, Moya W, Rodriguez O, Jijon A, Geipel A. Predictive value of angiogenic factors, clinical risk factors and uterine artery Doppler for pre-eclampsia and fetal growth restriction in second and third trimester pregnancies in an Ecuadorian population. J Matern Fetal Neonatal Med 2016;29:537-543.
  145. Kwiatkowski S, Bednarek-Jedrzejek M, Ksel J, et al. sFlt-1/PlGF and Doppler ultrasound parameters in SGA pregnancies with confirmed neonatal birth weight below 10th percentile. Pregnancy Hypertens 2018;14:79-85.
  146. Garcia-Manau P, Mendoza M, Bonacina E, et al. Soluble fms-like tyrosine kinase to placental growth factor ratio in different stages of early-onset fetal growth restriction and small for gestational age. Acta Obstet Gynecol Scand 2021;100:119-128.
  147. Chen W, Wei Q, Liang Q, Song S, Li J. Diagnostic capacity of sFlt-1/PlGF ratio in fetal growth restriction:A systematic review and meta-analysis. Placenta 2022;127:37-42.
  148. Cowans NJ, Spencer K. First-trimester ADAM12 and PAPP-A as markers for intrauterine fetal growth restriction through their roles in the insulin-like growth factor system. Prenat Diagn 2007;27:264-271.
  149. Stepan H, Kramer T, Faber R. Maternal plasma concentrations of soluble endoglin in pregnancies with intrauterine growth restriction. J Clin Endocrinol Metab 2007;92:2831-2834.
  150. Asvold BO, Vatten LJ, Romundstad PR, Jenum PA, Karumanchi SA, Eskild A. Angiogenic factors in maternal circulation and the risk of severe fetal growth restriction. Am J Epidemiol 2011;173:630-639.
  151. Nanjo S, Minami S, Mizoguchi M, et al. Levels of serum-circulating angiogenic factors within 1 week prior to delivery are closely related to conditions of pregnant women with pre-eclampsia, gestational hypertension, and/or fetal growth restriction. J Obstet Gynaecol Res 2017;43:1805-1814.
  152. Li Y, Liu J. MicroRNA-206 predicts raised fetal growth retardation risk through the interaction with vascular endothelial growth factor in pregnancies. Medicine (Baltimore) 2020;99:e18897.
  153. Tang Y, Ye W, Liu X, Lv Y, Yao C, Wei J. VEGF and sFLT-1 in serum of PIH patients and effects on the foetus. Exp Ther Med 2019;17:2123-2128.
  154. Bourque DK, Avila L, Penaherrera M, von Dadelszen P, Robinson WP. Decreased placental methylation at the H19/IGF2 imprinting control region is associated with normotensive intrauterine growth restriction but not preeclampsia. Placenta 2010;31:197-202.
  155. Qiu Q, Bell M, Lu X, et al. Significance of IGFBP-4 in the development of fetal growth restriction. J Clin Endocrinol Metab 2012;97:E1429-1439.
  156. Pang WW, Tsui MH, Sahota D, et al. A strategy for identifying circulating placental RNA markers for fetal growth assessment. Prenat Diagn 2009;29:495-504.
  157. Kaitu'u-Lino TJ, MacDonald TM, Cannon P, et al. Circulating SPINT1 is a biomarker of pregnancies with poor placental function and fetal growth restriction. Nat Commun 2020;11:2411.
  158. Kaitu'u-Lino TJ, Tong S, Walker SP, et al. Maternal circulating SPINT1 is reduced in small-for-gestational age pregnancies at 26 weeks:Growing up in Singapore towards health outcomes (GUSTO) cohort study. Placenta 2021;110:24-28.
  159. Murphy CN, Cluver CA, Walker SP, et al. Circulating SPINT1 Is Reduced in a Preeclamptic Cohort with Co-Existing Fetal Growth Restriction. J Clin Med 2022;11:901.
  160. MacDonald TM, Walker SP, Hiscock R, Cannon P, Harper A, Murray E. et al. Circulating Delta-like homolog 1 (DLK1) at 36 weeks is correlated with birthweight and is of placental origin. Placenta 2020;91:24-30.
  161. Krauss T, Emons G, Kuhn W, Augustin HG. Predictive value of routine circulating soluble endothelial cell adhesion molecule measurements during pregnancy. Clin Chem 2002;48:1418-1425.
  162. Schneuer FJ, Nassar N, Khambalia AZ, et al. First trimester screening of maternal placental protein 13 for predicting preeclampsia and small for gestational age:in-house study and systematic review. Placenta 2012;33:735-740.
  163. Spencer K, Yu CK, Cowans NJ, Otigbah C, Nicolaides KH. Prediction of pregnancy complications by first-trimester maternal serum PAPP-A and free beta-hCG and with second-trimester uterine artery Doppler. Prenat Diagn 2005;25:949-953.
  164. Gundu S, Kulkarni M, Gupte S, Gupte A, Gambhir M, Gambhir P. Correlation of first-trimester serum levels of pregnancy-associated plasma protein A with small-for-gestational-age neonates and preterm births. Int J Gynaecol Obstet 2016;133:159-163.
  165. Canini S, Prefumo F, Pastorino D, et al. Association between birth weight and first-trimester free beta-human chorionic gonadotropin and pregnancy-associated plasma protein A. Fertil Steril 2008;89:174-178.
  166. Crovetto F, Crispi F, Scazzocchio E, et al. First-trimester screening for early and late small-for-gestational-age neonates using maternal serum biochemistry, blood pressure and uterine artery Doppler. Ultrasound Obstet Gynecol 2014;43:34-40.
  167. Erez O, Romero R, Espinoza J, et al. The change in concentrations of angiogenic and anti-angiogenic factors in maternal plasma between the first and second trimesters in risk assessment for the subsequent development of preeclampsia and small-for-gestational age. J Matern Fetal Neonatal Med 2008;21:279-287.
  168. Regnault TR, Orbus RJ, de Vrijer B, et al. Placental expression of VEGF, PlGF and their receptors in a model of placental insufficiency-intrauterine growth restriction (PI-IUGR). Placenta 2002;23:132-144.
  169. Sifakis S, Akolekar R, Kappou D, Mantas N, Nicolaides KH. Maternal serum placental growth hormone at 11-13 weeks' gestation in pregnancies delivering small for gestational age neonates. J Matern Fetal Neonatal Med 2012;25:1796-1799.
  170. Cleaton MA, Dent CL, Howard M, et al. Fetus-derived DLK1 is required for maternal metabolic adaptations to pregnancy and is associated with fetal growth restriction. Nat Genet 2016;48:1473-1480.
  171. Pham A, Mitanchez D, Forhan A, et al. Low Maternal DLK1 Levels at 26 Weeks Is Associated With Small for Gestational Age at Birth. Front Endocrinol (Lausanne) 2022;13:836731.
  172. Ashraf UM, Hall DL, Rawls AZ, Alexander BT. Epigenetic processes during preeclampsia and effects on fetal development and chronic health. Clin Sci (Lond) 2021;135:2307-2327.
  173. Stepan H, Galindo A, Hund M, et al. Clinical utility of sFlt-1 and PlGF in screening, prediction, diagnosis and monitoring of pre-eclampsia and fetal growth restriction. Ultrasound Obstet Gynecol 2023;61:168-180.
  174. Guo Z, Yang F, Zhang J, et al. Whole-Genome Promoter Profiling of Plasma DNA Exhibits Diagnostic Value for Placenta-Origin Pregnancy Complications. Adv Sci (Weinh) 2020;7:1901819.
  175. Bahado-Singh R, Vishweswaraiah S, Mishra NK, Guda C, Radhakrishna U. Placental DNA methylation changes in detection of tetralogy of Fallot. Ultrasound Obstet Gynecol 2020;55:768-775.
  176. Liu J, Wu Y, Sun H, et al. Association between placental DNA methylation and fetal congenital heart disease. Mol Genet Genomics 2023;298:243-251.
  177. Liu J, Wu Y, Sun H, et al. Placental DNA Methylation Abnormalities in Prenatal Conotruncal Heart Defects. Front Genet 2022;13:878063.
  178. Zhu Y, Gomez JA, Laufer BI, Mordaunt CE, Mouat JS, Soto DC. et al. Placental methylome reveals a 22q13.33 brain regulatory gene locus associated with autism. Genome Biol 2022;23:46.
  179. Bahado-Singh RO, Vishweswaraiah S, Aydas B, Radhakrishna U. Placental DNA methylation changes and the early prediction of autism in full-term newborns. PLoS One 2021;16:e0253340.
  180. Appleton AA, Lester BM, Armstrong DA, Lesseur C, Marsit CJ. Examining the joint contribution of placental NR3C1 and HSD11B2 methylation for infant neurobehavior. Psychoneuroendocrinology 2015;52:32-42.
  181. Ursini G, Di Carlo P, Mukherjee S, et al. Prioritization of potential causative genes for schizophrenia in placenta. Nat Commun 2023;14:2613.
  182. Tehrani JM, Kennedy E, Tung PW, et al. Human placental microRNAs dysregulated by cadmium exposure predict neurobehavioral outcomes at birth. Pediatr Res 2023;93:1410-1418.
  183. Howerton CL, Morgan CP, Fischer DB, Bale TL. O-GlcNAc transferase (OGT) as a placental biomarker of maternal stress and reprogramming of CNS gene transcription in development. Proc Natl Acad Sci U S A 2013;110:5169-5174.
  184. Nugent BM, O'Donnell CM, Epperson CN, Bale TL. Placental H3K27me3 establishes female resilience to prenatal insults. Nat Commun 2018;9:2555.
  185. Chhabra D, Sharma S, Kho AT, et al. Fetal lung and placental methylation is associated with in utero nicotine exposure. Epigenetics 2014;9:1473-1484.
  186. Shorey-Kendrick LE, McEvoy CT, O'Sullivan SM, et al. Impact of vitamin C supplementation on placental DNA methylation changes related to maternal smoking:association with gene expression and respiratory outcomes. Clin Epigenetics 2021;13:177.
  187. Arendt LH, Ramlau-Hansen CH, Wilcox AJ, Henriksen TB, Olsen J, Lindhard MS. Placental Weight and Male Genital Anomalies:A Nationwide Danish Cohort Study. Am J Epidemiol 2016;183:1122-1128.
  188. Arrebola JP, Molina-Molina JM, Fernandez MF, et al. A novel biomarker for anti-androgenic activity in placenta reveals risks of urogenital malformations. Reproduction 2015;149:605-613.
  189. Fernandez MF, Olmos B, Granada A, et al. Human exposure to endocrine-disrupting chemicals and prenatal risk factors for cryptorchidism and hypospadias:a nested case-control study. Environ Health Perspect 2007;115:8-14.
  190. Green BB, Kappil M, Lambertini L, et al. Expression of imprinted genes in placenta is associated with infant neurobehavioral development. Epigenetics 2015;10:834-841.
  191. Lesseur C, Armstrong DA, Murphy MA, et al. Sex-specific associations between placental leptin promoter DNA methylation and infant neurobehavior. Psychoneuroendocrinology 2014;40:1-9.
  192. Redline RW, Minich N, Taylor HG, Hack M. Placental lesions as predictors of cerebral palsy and abnormal neurocognitive function at school age in extremely low birth weight infants (<1 kg). Pediatr Dev Pathol 2007;10:282-292.
  193. Maccani MA, Padbury JF, Lester BM, Knopik VS, Marsit CJ. Placental miRNA expression profiles are associated with measures of infant neurobehavioral outcomes. Pediatr Res 2013;74:272-278.
  194. Goodfellow LR, Cooper C, Harvey NC. Regulation of placental calcium transport and offspring bone health. Front Endocrinol (Lausanne) 2011;2:3.