1,022
Views
13
CrossRef citations to date
0
Altmetric
Original Article

Maternal plasma soluble TRAIL is decreased in preeclampsia

, , , , , , , , & show all
Pages 217-227 | Received 16 Apr 2013, Accepted 16 May 2013, Published online: 13 Aug 2013

References

  • Romero R, Lockwood C, Oyarzun E, Hobbins JC. Toxemia: new concepts in an old disease. Semin Perinatol 1988;12:302–23
  • Romero R. Prenatal medicine: the child is the father of the man. 1996. J Matern Fetal Neonatal Med 2009;22:636–9
  • Redman CW. Preeclampsia: a multi-stress disorder. Rev Med Interne 2011;32:S41–44
  • Roberts JM, Gammill HS. Preeclampsia: recent insights. Hypertension 2005;46:1243–9
  • Sibai BM. Evaluation and management of severe preeclampsia before 34 weeks' gestation. Am J Obstet Gynecol 2011;205:191–8
  • Barton JR, Sibai BM. Acute life-threatening emergencies in preeclampsia–eclampsia. Clin Obstet Gynecol 1992;35:402–13
  • Than NG, Romero R, Hillermann R, et al. Prediction of preeclampsia - a workshop report. Placenta 2008;29:S83–5
  • Romero R, Vizoso J, Emamian M, et al. Clinical significance of liver dysfunction in pregnancy-induced hypertension. Am J Perinatol 1988;5:146–51
  • Di Renzo GC. The great obstetrical syndromes. J Matern Fetal Neonatal Med 2009;22:633–5
  • von Dadelszen P, Magee LA. Hypertensive disease in pregnancy. Preface. Best Pract Res Clin Obstet Gynaecol 2011;25:389–90
  • Romero R, Duffy TP. Platelet disorders in pregnancy. Clin Perinatol 1980;7:327–48
  • Redman CW, Sargent IL. Latest advances in understanding preeclampsia. Science 2005;308:1592–4
  • Redman CW, Sargent IL. Preeclampsia and the systemic inflammatory response. Semin Nephrol 2004;24:565–70
  • Payne B, Magee LA, von Dadelszen P. Assessment, surveillance and prognosis in pre-eclampsia. Best Pract Res Clin Obstet Gynaecol 2011;25:449–62
  • Dietl J. The pathogenesis of pre-eclampsia: new aspects. J Perinat Med 2000;28:464–71
  • Steegers EA, von Dadelszen P, Duvekot JJ, Pijnenborg R. Pre-eclampsia. Lancet 2010;376:631–44
  • Hofbauer J. The evolution of the biologic concept of the etiology of late toxemia. Am J Obstet Gynecol 1946;51:514–18
  • Theobald GW. The toxaemias of pregnancy. J Obstet Gynaecol Br Emp 1946;53:17–41
  • Page NM, Lowry PJ. Is ‘pre-eclampsia' simply a response to the side effects of a placental tachykinin? J Endocrinol 2000;167:355–61
  • Page NM, Woods RJ, Gardiner SM, et al. Excessive placental secretion of neurokinin B during the third trimester causes pre-eclampsia. Nature 2000;405:797–800
  • Brosens I, Pijnenborg R, Vercruysse L, Romero R. The “Great Obstetrical Syndromes” are associated with disorders of deep placentation. Am J Obstet Gynecol 2011;204:193–201
  • Pijnenborg R, Brosens I, Romero R. Placental bed disorders. United States of America: Cambridge University Press; 2010
  • Brosens IA. Morphological changes in the utero-placental bed in pregnancy hypertension. Clin Obstet Gynaecol 1977;4:573–93
  • Brosens JJ, Pijnenborg R, Brosens IA. The myometrial junctional zone spiral arteries in normal and abnormal pregnancies: a review of the literature. Am J Obstet Gynecol 2002;187:1416–23
  • Pijnenborg R, Anthony J, Davey DA, et al. Placental bed spiral arteries in the hypertensive disorders of pregnancy. Br J Obstet Gynaecol 1991;98:648–55
  • Robertson WB, Brosens I, Dixon G. Maternal uterine vascular lesions in the hypertensive complications of pregnancy. Perspect Nephrol Hypertens 1976;5:115–27
  • Ogge G, Chaiworapongsa T, Romero R, et al. Placental lesions associated with maternal underperfusion are more frequent in early-onset than in late-onset preeclampsia. J Perinat Med 2011;39:641–52
  • George EM, Granger JP. Linking placental ischemia and hypertension in preeclampsia: role of endothelin 1. Hypertension 2012;60:507–11
  • Gilbert JS, Ryan MJ, LaMarca BB, et al. Pathophysiology of hypertension during preeclampsia: linking placental ischemia with endothelial dysfunction. Am J Physiol Heart Circ Physiol 2008;294:H541–50
  • Sacks GP, Studena K, Sargent K, Redman CW. Normal pregnancy and preeclampsia both produce inflammatory changes in peripheral blood leukocytes akin to those of sepsis. Am J Obstet Gynecol 1998;179:80–6
  • Redman CW, Sacks GP, Sargent IL. Preeclampsia: an excessive maternal inflammatory response to pregnancy. Am J Obstet Gynecol 1999;180:499–506
  • Gervasi MT, Chaiworapongsa T, Pacora P, et al. Phenotypic and metabolic characteristics of monocytes and granulocytes in preeclampsia. Am J Obstet Gynecol 2001;185:792–7
  • Cindrova-Davies T. Gabor Than Award Lecture 2008: pre-eclampsia - from placental oxidative stress to maternal endothelial dysfunction. Placenta 2009;30:S55–65
  • Friedman SA, Schiff E, Emeis JJ, et al. Biochemical corroboration of endothelial involvement in severe preeclampsia. Am J Obstet Gynecol 1995;172:202–3
  • Lyall F, Greer IA. The vascular endothelium in normal pregnancy and pre-eclampsia. Rev Reprod 1996;1:107–16
  • Roberts JM, Hubel CA. The two stage model of preeclampsia: variations on the theme. Placenta 2009;30:S32–7
  • Roberts JM, Taylor RN, Musci TJ, et al. Preeclampsia: an endothelial cell disorder. Am J Obstet Gynecol 1989;161:1200–4
  • Roberts JM, Taylor RN, Goldfien A. Clinical and biochemical evidence of endothelial cell dysfunction in the pregnancy syndrome preeclampsia. Am J Hypertens 1991;4:700–8
  • Roberts JM. Endothelial dysfunction in preeclampsia. Semin Reprod Endocrinol 1998;16:5–15
  • Taylor RN, de Groot CJ, Cho YK, Lim KH. Circulating factors as markers and mediators of endothelial cell dysfunction in preeclampsia. Semin Reprod Endocrinol 1998;16:17–31
  • Chaiworapongsa T, Romero R, Yoshimatsu J, et al. Soluble adhesion molecule profile in normal pregnancy and pre-eclampsia. J Matern Fetal Neonatal Med 2002;12:19–27
  • Petrozella L, Mahendroo M, Timmons B, et al. Endothelial microparticles and the antiangiogenic state in preeclampsia and the postpartum period. Am J Obstet Gynecol 2012;207:140 e120–46
  • Dekker GA, van Geijn HP. Endothelial dysfunction in preeclampsia. Part II: reducing the adverse consequences of endothelial cell dysfunction in preeclampsia; therapeutic perspectives. J Perinat Med 1996;24:119–39
  • Dekker GA, van Geijn HP. Endothelial dysfunction in preeclampsia. Part I: primary prevention. Therapeutic perspectives. J Perinat Med 1996;24:99–117
  • Gervasi MT, Chaiworapongsa T, Naccasha N, et al. Phenotypic and metabolic characteristics of maternal monocytes and granulocytes in preterm labor with intact membranes. Am J Obstet Gynecol 2001;185:1124–9
  • Gotsch F, Romero R, Erez O, et al. The preterm parturition syndrome and its implications for understanding the biology, risk assessment, diagnosis, treatment and prevention of preterm birth. J Matern Fetal Neonatal Med 2009;22:5–23
  • Gotsch F, Romero R, Kusanovic JP, et al. The anti-inflammatory limb of the immune response in preterm labor, intra-amniotic infection/inflammation, and spontaneous parturition at term: a role for interleukin-10. J Matern Fetal Neonatal Med 2008;21:529–47
  • Gervasi MT, Chaiworapongsa T, Naccasha N, et al. Maternal intravascular inflammation in preterm premature rupture of membranes. J Matern Fetal Neonatal Med 2002;11:171–5
  • Loukovaara MJ, Alfthan HV, Kurki MT, et al. Serum highly sensitive C-reactive protein in preterm premature rupture of membranes. Eur J Obstet Gynecol Reprod Biol 2003;110:26–8
  • Ogge G, Romero R, Chaiworapongsa T, et al. Leukocytes of pregnant women with small-for-gestational age neonates have a different phenotypic and metabolic activity from those of women with preeclampsia. J Matern Fetal Neonatal Med 2010;23:476–87
  • Kusanovic JP, Romero R, Hassan SS, et al. Maternal serum soluble CD30 is increased in normal pregnancy, but decreased in preeclampsia and small for gestational age pregnancies. J Matern Fetal Neonatal Med 2007;20:867–78
  • Labarrere C, Manni J, Salas P, Althabe O. Intrauterine growth retardation of unknown etiology. I. Serum complement and circulating immune complexes in mothers and infants. Am J Reprod Immunol Microbiol 1985;8:87–93
  • Labarrere CA, Althabe OH. Intrauterine growth retardation of unknown etiology: II. Serum complement and circulating immune complexes in maternal sera and their relationship with parity and chronic villitis. Am J Reprod Immunol Microbiol 1986;12:4–6
  • Johnston TA, Greer IA, Dawes J, Calder AA. Neutrophil activation in small for gestational age pregnancies. Br J Obstet Gynaecol 1991;98:105–6
  • Kobayashi N, Yamada H, Kishida T, et al. Hypocomplementemia correlates with intrauterine growth retardation in systemic lupus erythematosus. Am J Reprod Immunol 1999;42:153–9
  • Sabatier F, Bretelle F, D'Ercole C, et al. Neutrophil activation in preeclampsia and isolated intrauterine growth restriction. Am J Obstet Gynecol 2000;183:1558–63
  • Johnson MR, Anim-Nyame N, Johnson P, et al. Does endothelial cell activation occur with intrauterine growth restriction? BJOG 2002;109:836–9
  • Coata G, Pennacchi L, Bini V, et al. Soluble adhesion molecules: marker of pre-eclampsia and intrauterine growth restriction. J Matern Fetal Neonatal Med 2002;12:28–34
  • Tjoa ML, van Vugt JM, Go AT, et al. Elevated C-reactive protein levels during first trimester of pregnancy are indicative of preeclampsia and intrauterine growth restriction. J Reprod Immunol 2003;59:29–37
  • Cetin I, Cozzi V, Pasqualini F, et al. Elevated maternal levels of the long pentraxin 3 (PTX3) in preeclampsia and intrauterine growth restriction. Am J Obstet Gynecol 2006;194:1347–53
  • Girardi G, Yarilin D, Thurman JM, et al. Complement activation induces dysregulation of angiogenic factors and causes fetal rejection and growth restriction. J Exp Med 2006;203:2165–75
  • Laskowska M, Laskowska K, Leszczynska-Gorzelak B, Oleszczuk J. Comparative analysis of the maternal and umbilical interleukin-8 levels in normal pregnancies and in pregnancies complicated by preeclampsia with intrauterine normal growth and intrauterine growth retardation. J Matern Fetal Neonatal Med 2007;20:527–32
  • Laskowska M, Laskowska K, Leszczynska-Gorzelak B, Oleszczuk J. Maternal and umbilical sTNF-R1 in preeclamptic pregnancies with intrauterine normal and growth retarded fetus. Hypertens Pregn 2007;26:13–21
  • Erez O, Romero R, Hoppensteadt D, et al. Tissue factor and its natural inhibitor in pre-eclampsia and SGA. J Matern Fetal Neonatal Med 2008;21:855–69
  • Kim YM, Chaiworapongsa T, Gomez Rc. Failure of physiologic transformation of the spiral arteries in the placental bed in preterm premature rupture of membranes. Am J Obstet Gynecol 2002;187:1137–42
  • Kim YM, Bujold E, Chaiworapongsa T, et al. Failure of physiologic transformation of the spiral arteries in patients with preterm labor and intact membranes. Am J Obstet Gynecol 2003;189:1063–9
  • Brosens I, Dixon HG, Robertson WB. Fetal growth retardation and the arteries of the placental bed. Br J Obstet Gynaecol 1977;84:656–63
  • Torry DS, Wang HS, Wang TH, et al. Preeclampsia is associated with reduced serum levels of placenta growth factor. Am J Obstet Gynecol 1998;179:1539–44
  • Tidwell SC, Ho HN, Chiu WH, et al. Low maternal serum levels of placenta growth factor as an antecedent of clinical preeclampsia. Am J Obstet Gynecol 2001;184:1267–72
  • Taylor RN, Grimwood J, Taylor RS, et al. Longitudinal serum concentrations of placental growth factor: evidence for abnormal placental angiogenesis in pathologic pregnancies. Am J Obstet Gynecol 2003;188:177–82
  • Reuvekamp A, Velsing-Aarts FV, Poulina IE, et al. Selective deficit of angiogenic growth factors characterises pregnancies complicated by pre-eclampsia. Br J Obstet Gynaecol 1999;106:1019–22
  • Crispi F, Dominguez C, Llurba E, et al. Placental angiogenic growth factors and uterine artery Doppler findings for characterization of different subsets in preeclampsia and in isolated intrauterine growth restriction. Am J Obstet Gynecol 2006;195:201–7
  • Chaiworapongsa T, Romero R, Gotsch F, et al. Low maternal concentrations of soluble vascular endothelial growth factor receptor-2 in preeclampsia and small for gestational age. J Matern Fetal Neonatal Med 2008;21:41–52
  • Garovic VD. The role of angiogenic factors in the prediction and diagnosis of preeclampsia superimposed on chronic hypertension. Hypertension 2012;59:555–7
  • Ghosh SK, Raheja S, Tuli A, et al. Serum PLGF as a potential biomarker for predicting the onset of preeclampsia. Arch Gynecol Obstet 2012;285:417–22
  • Hagmann H, Thadhani R, Benzing T, et al. The promise of angiogenic markers for the early diagnosis and prediction of preeclampsia. Clin Chem 2012;58:837–45
  • Molvarec A, Szarka A, Walentin S, et al. Circulating angiogenic factors determined by electrochemiluminescence immunoassay in relation to the clinical features and laboratory parameters in women with pre-eclampsia. Hypertens Res 2010;33:892–8
  • Chaiworapongsa T, Romero R, Tarca AL, et al. A decrease in maternal plasma concentrations of sVEGFR-2 precedes the clinical diagnosis of preeclampsia. Am J Obstet Gynecol 2010;202:550.e1–10
  • Chaiworapongsa T, Romero R, Savasan ZA, et al. Maternal plasma concentrations of angiogenic/anti-angiogenic factors are of prognostic value in patients presenting to the obstetrical triage area with the suspicion of preeclampsia. J Matern Fetal Neonatal Med 2011;24:1187–207
  • Staff AC, Braekke K, Johnsen GM, et al. Circulating concentrations of soluble endoglin (CD105) in fetal and maternal serum and in amniotic fluid in preeclampsia. Am J Obstet Gynecol 2007;197:176 e171–76
  • Lam C, Lim KH, Karumanchi SA. Circulating angiogenic factors in the pathogenesis and prediction of preeclampsia. Hypertension 2005;46:1077–85
  • Chaiworapongsa T, Romero R, Kim YM, et al. Plasma soluble vascular endothelial growth factor receptor-1 concentration is elevated prior to the clinical diagnosis of pre-eclampsia. J Matern Fetal Neonatal Med 2005;17:3–18
  • Moore Simas TA, Crawford SL, Solitro MJ, et al. Angiogenic factors for the prediction of preeclampsia in high-risk women. Am J Obstet Gynecol 2007;197:244 e241–8
  • 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;21:279–87
  • Romero R, Nien JK, Espinoza J, et al. A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small for gestational age neonate. J Matern Fetal Neonatal Med 2008;21:9–23
  • Unal ER, Robinson CJ, Johnson DD, Chang EY. Second-trimester angiogenic factors as biomarkers for future-onset preeclampsia. Am J Obstet Gynecol 2007;197:211 e211–14
  • Widmer M, Villar J, Benigni A, et al. Mapping the theories of preeclampsia and the role of angiogenic factors: a systematic review. Obstet Gynecol 2007;109:168–80
  • Park CW, Park JS, Shim SS, et al. An elevated maternal plasma, but not amniotic fluid, soluble fms-like tyrosine kinase-1 (sFlt-1) at the time of mid-trimester genetic amniocentesis is a risk factor for preeclampsia. Am J Obstet Gynecol 2005;193:984–9
  • Chaiworapongsa T, Romero R, Espinoza J, et al. Evidence supporting a role for blockade of the vascular endothelial growth factor system in the pathophysiology of preeclampsia. Young Investigator Award. Am J Obstet Gynecol 2004;190:1541–7; discussion 1547–50
  • Maynard SE, Min JY, Merchan J, et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest 2003;111:649–58
  • Koga K, Osuga Y, Yoshino O, et al. Elevated serum soluble vascular endothelial growth factor receptor 1 (sVEGFR-1) levels in women with preeclampsia. J Clin Endocrinol Metab 2003;88:2348–51
  • Tsatsaris V, Goffin F, Munaut C, et al. Overexpression of the soluble vascular endothelial growth factor receptor in preeclamptic patients: pathophysiological consequences. J Clin Endocrinol Metab 2003;88:5555–63
  • Venkatesha S, Toporsian M, Lam C, et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med 2006;12:642–9
  • Luttun A, Carmeliet P. Soluble VEGF receptor Flt1: the elusive preeclampsia factor discovered? J Clin Invest 2003;111:600–2
  • Bujold E, Romero R, Chaiworapongsa T, et al. Evidence supporting that the excess of the sVEGFR-1 concentration in maternal plasma in preeclampsia has a uterine origin. J Matern Fetal Neonatal Med 2005;18:9–16
  • Rana S, Hacker MR, Modest AM, et al. Circulating angiogenic factors and risk of adverse maternal and perinatal outcomes in twin pregnancies with suspected preeclampsia. Hypertension 2012;60:451–8
  • Rana S, Cerdeira AS, Wenger J, et al. Plasma concentrations of soluble endoglin versus standard evaluation in patients with suspected preeclampsia. PLoS One 2012;7:e48259
  • Moore AG, Young H, Keller JM, et al. Angiogenic biomarkers for prediction of maternal and neonatal complications in suspected preeclampsia. J Matern Fetal Neonatal Med 2012;25:2651--7
  • Bellamy L, Casas JP, Hingorani AD, Williams DJ. Pre-eclampsia and risk of cardiovascular disease and cancer in later life: systematic review and meta-analysis. Br Med J 2007;335:1--12
  • Jacquemyn Y, Jochems L, Duiker E, et al. Long-term renal function after HELLP syndrome. Gynecol Obstet Invest 2004;57:117–20
  • Sibai BM, Mercer B, Sarinoglu C. Severe preeclampsia in the second trimester: recurrence risk and long-term prognosis. Am J Obstet Gynecol 1991;165:1408–12
  • Suzuki H, Watanabe Y, Arima H, et al. Short- and long-term prognosis of blood pressure and kidney disease in women with a past history of preeclampsia. Clin Exp Nephrol 2008;12:102–9
  • Hannaford P, Ferry S, Hirsch S. Cardiovascular sequelae of toxaemia of pregnancy. Heart 1997;77:154–8
  • McDonald SD, Malinowski A, Zhou Q, et al. Cardiovascular sequelae of preeclampsia/eclampsia: a systematic review and meta-analyses. Am Heart J 2008;156:918–30
  • Cassidy-Bushrow AE, Bielak LF, Rule AD, et al. Hypertension during pregnancy is associated with coronary artery calcium independent of renal function. J Womens Health (Larchmt) 2009;18:1709–16
  • Lykke JA, Langhoff-Roos J, Sibai BM, et al. Hypertensive pregnancy disorders and subsequent cardiovascular morbidity and type 2 diabetes mellitus in the mother. Hypertension 2009;53:944–51
  • Garovic VD, Bailey KR, Boerwinkle E, et al. Hypertension in pregnancy as a risk factor for cardiovascular disease later in life. J Hypertens 2010;28:826–33
  • Mongraw-Chaffin ML, Cirillo PM, Cohn BA. Preeclampsia and cardiovascular disease death: prospective evidence from the child health and development studies cohort. Hypertension 2010;56:166–71
  • Lin YS, Tang CH, Yang CY, et al. Effect of pre-eclampsia-eclampsia on major cardiovascular events among peripartum women in Taiwan. Am J Cardiol 2011;107:325–30
  • Newstead J, von Dadelszen P, Magee LA. Preeclampsia and future cardiovascular risk. Expert Rev Cardiovasc Ther 2007;5:283–94
  • Rodie VA, Freeman DJ, Sattar N, Greer IA. Pre-eclampsia and cardiovascular disease: metabolic syndrome of pregnancy? Atherosclerosis 2004;175:189–202
  • Potter JM, Nestel PJ. The hyperlipidemia of pregnancy in normal and complicated pregnancies. Am J Obstet Gynecol 1979;133:165–70
  • Branch DW, Mitchell MD, Miller E, et al. Pre-eclampsia and serum antibodies to oxidised low-density lipoprotein. Lancet 1994;343:645–6
  • Hubel CA, McLaughlin MK, Evans RW, et al. Fasting serum triglycerides, free fatty acids, and malondialdehyde are increased in preeclampsia, are positively correlated, and decrease within 48 hours post partum. Am J Obstet Gynecol 1996;174:975–82
  • Ogura K, Miyatake T, Fukui O, et al. Low-density lipoprotein particle diameter in normal pregnancy and preeclampsia. J Atheroscler Thromb 2002;9:42–7
  • Kaaja R, Tikkanen MJ, Viinikka L, Ylikorkala O. Serum lipoproteins, insulin, and urinary prostanoid metabolites in normal and hypertensive pregnant women. Obstet Gynecol 1995;85:353–6
  • Hermes W, Van Kesteren F, De Groot CJ. Preeclampsia and cardiovascular risk. Minerva Ginecol 2012;64:281–92
  • Girouard J, Giguere Y, Moutquin JM, Forest JC. Previous hypertensive disease of pregnancy is associated with alterations of markers of insulin resistance. Hypertension 2007;49:1056–62
  • Kaaja R. Insulin resistance syndrome in preeclampsia. Semin Reprod Endocrinol 1998;16:41–6
  • Kaaja R, Laivuori H, Laakso M, et al. Evidence of a state of increased insulin resistance in preeclampsia. Metabolism 1999;48:892–6
  • Dietl J. Maternal obesity and complications during pregnancy. J Perinat Med 2005;33:100–5
  • Hirschfield GM, Pepys MB. C-reactive protein and cardiovascular disease: new insights from an old molecule. QJM 2003;96:793–807
  • Black S, Kushner I, Samols D. C-reactive Protein. J Biol Chem 2004;279:48487–90
  • Pepys MB, Hirschfield GM. C-reactive protein: a critical update. J Clin Invest 2003;111:1805–12
  • Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation 2000;102:2165–8
  • Calabro P, Willerson JT, Yeh ET. Inflammatory cytokines stimulated C-reactive protein production by human coronary artery smooth muscle cells. Circulation 2003;108:1930–2
  • Zhang YX, Cliff WJ, Schoefl GI, Higgins G. Coronary C-reactive protein distribution: its relation to development of atherosclerosis. Atherosclerosis 1999;145:375–9
  • Scirica BM, Morrow DA. Is C-reactive protein an innocent bystander or proatherogenic culprit? The verdict is still out. Circulation 2006;113:2128–34; discussion 2151
  • Verma S, Devaraj S, Jialal I. Is C-reactive protein an innocent bystander or proatherogenic culprit? C-reactive protein promotes atherothrombosis. Circulation 2006;113:2135–50; discussion 2150
  • de Beer FC, Soutar AK, Baltz ML, et al. Low density lipoprotein and very low density lipoprotein are selectively bound by aggregated C-reactive protein. J Exp Med 1982;156:230–42
  • Pepys MB, Rowe IF, Baltz ML. C-reactive protein: binding to lipids and lipoproteins. Int Rev Exp Pathol 1985;27:83–111
  • Kaptoge S, Di Angelantonio E, Lowe G, et al. C-reactive protein concentration and risk of coronary heart disease, stroke, and mortality: an individual participant meta-analysis. Lancet 2010;375:132–40
  • Correia M, Mendes S, Araujo C, et al. Subclavian steal syndrome in a coronary patient. Rev Port Cardiol 2011;30:633–5
  • Danesh J, Wheeler JG, Hirschfield GM, et al. C-reactive protein and other circulating markers of inflammation in the prediction of coronary heart disease. N Engl J Med 2004;350:1387–97
  • Ridker PM, Buring JE, Shih J, et al. Prospective study of C-reactive protein and the risk of future cardiovascular events among apparently healthy women. Circulation 1998;98:731–3
  • Sakkinen P, Abbott RD, Curb JD, et al. C-reactive protein and myocardial infarction. J Clin Epidemiol 2002;55:445–51
  • Koenig W, Sund M, Frohlich M, et al. C-Reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: results from the MONICA (Monitoring Trends and Determinants in Cardiovascular Disease) Augsburg Cohort Study, 1984 to 1992. Circulation 1999;99:237–42
  • Casas JP, Shah T, Cooper J, et al. Insight into the nature of the CRP-coronary event association using Mendelian randomization. Int J Epidemiol 2006;35:922–31
  • Wilson PW, Nam BH, Pencina M, et al. C-reactive protein and risk of cardiovascular disease in men and women from the Framingham Heart Study. Arch Intern Med 2005;165:2473–8
  • Pearson TA, Mensah GA, Hong Y, Smith SC, Jr CDC/AHA Workshop on Markers of Inflammation and Cardiovascular Disease: application to clinical and public health practice: overview. Circulation 2004;110:e543–4
  • Greenland P, Alpert JS, Beller GA, et al. 2010 ACCF/AHA guideline for assessment of cardiovascular risk in asymptomatic adults: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation 2010;122:e584–636
  • Madjid M, Willerson JT. Inflammatory markers in coronary heart disease. Br Med Bull 2011;100:23–38
  • Correia LC, Esteves JP. C-Reactive protein and outcomes in acute coronary syndromes: a systematic review and meta-analysis. Arq Bras Cardiol 2011;97:76–85
  • Abd TT, Eapen DJ, Bajpai A, et al. The role of C-reactive protein as a risk predictor of coronary atherosclerosis: implications from the JUPITER trial. Curr Atheroscler Rep 2011;13:154–61
  • Rietzschel E, De Buyzere M. High-sensitive C-reactive protein: universal prognostic and causative biomarker in heart disease? Biomark Med 2012;6:19–34
  • Rebelo F, Schlussel MM, Vaz JS, et al. C-reactive protein and later preeclampsia: systematic review and meta-analysis taking into account the weight status. Journal of hypertension 2013;31:16–26
  • Garcia RG, Celedon J, Sierra-Laguado J, et al. Raised C-reactive protein and impaired flow-mediated vasodilation precede the development of preeclampsia. Am J Hypertens 2007;20:98–103
  • Qiu C, Luthy DA, Zhang C, Walsh SW, et al. A prospective study of maternal serum C-reactive protein concentrations and risk of preeclampsia. Am J Hypertens 2004;17:154–60
  • de Jonge LL, Steegers EA, Ernst GD, et al. C-reactive protein levels, blood pressure and the risks of gestational hypertensive complications: the Generation R Study. J Hypertens 2011;29:2413–21
  • Gandevani SB, Banaem LM, Mohamadi B, et al. Association of high-sensitivity C-reactive protein serum levels in early pregnancy with the severity of preeclampsia and fetal birth weight. J Perinat Med 2012;0:1–5
  • Kucukgoz Gulec U, Tuncay Ozgunen F, Baris Guzel A, et al. An analysis of C-reactive protein, procalcitonin, and D-dimer in pre-eclamptic patients. Am J Reprod Immunol 2012;68:331–7
  • Cemgil Arikan D, Aral M, Coskun A, Ozer A. Plasma IL-4, IL-8, IL-12, interferon-gamma and CRP levels in pregnant women with preeclampsia, and their relation with severity of disease and fetal birth weight. The journal of maternal-fetal & neonatal medicine: the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstetricians 2012;25:1569–73
  • Teran E, Escudero C, Calle A. C-reactive protein during normal pregnancy and preeclampsia. Int J Gynaecol Obstet 2005;89:299–300
  • Ertas IE, Kahyaoglu S, Yilmaz B, et al. Association of maternal serum high sensitive C-reactive protein level with body mass index and severity of pre-eclampsia at third trimester. J Obstet Gynaecol Res 2010;36:970–7
  • Paternoster DM, Fantinato S, Stella A, et al. C-reactive protein in hypertensive disorders in pregnancy. Clinical and applied thrombosis/hemostasis: official journal of the International Academy of Clinical and Applied Thrombosis/Hemostasis 2006;12:330–7
  • Wiley SR, Schooley K, Smolak PJ, et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity 1995;3:673–82
  • Pitti RM, Marsters SA, Ruppert S, et al. Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem 1996;271:12687–90
  • Kavurma MM, Schoppet M, Bobryshev YV, et al. TRAIL stimulates proliferation of vascular smooth muscle cells via activation of NF-kappaB and induction of insulin-like growth factor-1 receptor. J Biol Chem 2008;283:7754–62
  • Zauli G, Pandolfi A, Gonelli A, et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) sequentially upregulates nitric oxide and prostanoid production in primary human endothelial cells. Circ Res 2003;92:732–40
  • Di Pietro R, Mariggio MA, Guarnieri S, et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) regulates endothelial nitric oxide synthase (eNOS) activity and its localization within the human vein endothelial cells (HUVEC) in culture. J Cell Biochem 2006;97:782–94
  • Secchiero P, Gonelli A, Carnevale E, et al. TRAIL promotes the survival and proliferation of primary human vascular endothelial cells by activating the Akt and ERK pathways. Circulation 2003;107:2250–6
  • Secchiero P, Corallini F, di Iasio MG, et al. TRAIL counteracts the proadhesive activity of inflammatory cytokines in endothelial cells by down-modulating CCL8 and CXCL10 chemokine expression and release. Blood 2005;105:3413–19
  • Michowitz Y, Goldstein E, Roth A, et al. The involvement of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in atherosclerosis. J Am Coll Cardiol 2005;45:1018–24
  • Renshaw SA, Parmar JS, Singleton V, et al. Acceleration of human neutrophil apoptosis by TRAIL. J Immunol 2003;170:1027–33
  • Cziupka K, Busemann A, Partecke LI, et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) improves the innate immune response and enhances survival in murine polymicrobial sepsis. Crit Care Med 2010;38:2169–74
  • McGrath EE, Marriott HM, Lawrie A, et al. TNF-related apoptosis-inducing ligand (TRAIL) regulates inflammatory neutrophil apoptosis and enhances resolution of inflammation. J Leukoc Biol 2011;90:855–65
  • Deftereos S, Giannopoulos G, Kossyvakis C, et al. Association of soluble tumour necrosis factor-related apoptosis-inducing ligand levels with coronary plaque burden and composition. Heart 2012;98:214–18
  • Volpato S, Ferrucci L, Secchiero P, et al. Association of tumor necrosis factor-related apoptosis-inducing ligand with total and cardiovascular mortality in older adults. Atherosclerosis 2011;215:452–8
  • Mori K, Ikari Y, Jono S, et al. Association of serum TRAIL level with coronary artery disease. Thromb Res 2010;125:322–5
  • Secchiero P, Corallini F, Ceconi C, et al. Potential prognostic significance of decreased serum levels of TRAIL after acute myocardial infarction. PLoS One 2009;4:e4442
  • Zauli G, Monasta L, Vecchi Brumatti L, et al. The early determination of circulating TRAIL levels does not predict the development of pre-eclampsia. Placenta 2012;33:135–6
  • Sibai BM, Ewell M, Levine RJ, et al. Risk factors associated with preeclampsia in healthy nulliparous women. The Calcium for Preeclampsia Prevention (CPEP) Study Group. Am J Obstet Gynecol 1997;177:1003–10
  • Alexander GR, Himes JH, Kaufman RB, et al. A United States national reference for fetal growth. Obstet Gynecol 1996;87:163–8
  • Seeds JW. Impaired fetal growth: definition and clinical diagnosis. Obstet Gynecol 1984;64:303–10
  • Pan G, O'Rourke K, Chinnaiyan AM, et al. The receptor for the cytotoxic ligand TRAIL. Science 1997;276:111–13
  • Pan G, Ni J, Wei YF, et al. An antagonist decoy receptor and a death domain-containing receptor for TRAIL. Science 1997;277:815–18
  • Schneider P, Bodmer JL, Thome M, et al. Characterization of two receptors for TRAIL. FEBS Lett 1997;416:329–34
  • Chaudhary PM, Eby M, Jasmin A, et al. Death receptor 5, a new member of the TNFR family, and DR4 induce FADD-dependent apoptosis and activate the NF-kappaB pathway. Immunity 1997;7:821–30
  • Sheridan JP, Marsters SA, Pitti RM, et al. Control of TRAIL-induced apoptosis by a family of signaling and decoy receptors. Science 1997;277:818–21
  • Bodmer JL, Holler N, Reynard S, et al. TRAIL receptor-2 signals apoptosis through FADD and caspase-8. Nat Cell Biol 2000;2:241–3
  • Kischkel FC, Lawrence DA, Chuntharapai A, et al. Apo2L/TRAIL-dependent recruitment of endogenous FADD and caspase-8 to death receptors 4 and 5. Immunity 2000;12:611–20
  • Kischkel FC, Hellbardt S, Behrmann I, et al. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J 1995;14:5579–88
  • Sprick MR, Weigand MA, Rieser E, et al. FADD/MORT1 and caspase-8 are recruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated by TRAIL receptor 2. Immunity 2000;12:599–609
  • Bouillet P, Strasser A. BH3-only proteins - evolutionarily conserved proapoptotic Bcl-2 family members essential for initiating programmed cell death. J Cell Sci 2002;115:1567–74
  • Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 1998;94:491–501
  • Degli-Esposti MA, Smolak PJ, Walczak H, et al. Cloning and characterization of TRAIL-R3, a novel member of the emerging TRAIL receptor family. J Exp Med 1997;186:1165–70
  • Marsters SA, Sheridan JP, Pitti RM, et al. A novel receptor for Apo2L/TRAIL contains a truncated death domain. Curr Biol 1997;7:1003–6
  • Simonet WS, Lacey DL, Dunstan CR, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997;89:309–19
  • Emery JG, McDonnell P, Burke MB, et al. Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL. J Biol Chem 1998;273:14363–7
  • Wajant H, Moosmayer D, Wuest T, et al. Differential activation of TRAIL-R1 and -2 by soluble and membrane TRAIL allows selective surface antigen-directed activation of TRAIL-R2 by a soluble TRAIL derivative. Oncogene 2001;20:4101–6
  • Tecchio C, Huber V, Scapini P, et al. IFNalpha-stimulated neutrophils and monocytes release a soluble form of TNF-related apoptosis-inducing ligand (TRAIL/Apo-2 ligand) displaying apoptotic activity on leukemic cells. Blood 2004;103:3837–44
  • Kaplan MJ, Ray D, Mo RR, et al. TRAIL (Apo2 ligand) and TWEAK (Apo3 ligand) mediate CD4+ T cell killing of antigen-presenting macrophages. J Immunol 2000;164:2897–904
  • Chen L, Liu X, Zhu Y, et al. Localization and variation of TRAIL and its receptors in human placenta during gestation. Life Sci 2004;74:1479–86
  • Phillips TA, Ni J, Pan G, et al. TRAIL (Apo-2L) and TRAIL receptors in human placentas: implications for immune privilege. J Immunol 1999;162:6053–9
  • Keogh RJ, Harris LK, Freeman A, et al. Fetal-derived trophoblast use the apoptotic cytokine tumor necrosis factor-alpha-related apoptosis-inducing ligand to induce smooth muscle cell death. Circ Res 2007;100:834–41
  • Whitley GS, Cartwright JE. Trophoblast-mediated spiral artery remodelling: a role for apoptosis. J Anat 2009;215:21–6
  • Jimenez MF, Watson RW, Parodo J, et al. Dysregulated expression of neutrophil apoptosis in the systemic inflammatory response syndrome. Arch Surg 1997;132:1263–9; discussion 1269–70
  • Secchiero P, Candido R, Corallini F, et al. Systemic tumor necrosis factor-related apoptosis-inducing ligand delivery shows antiatherosclerotic activity in apolipoprotein E-null diabetic mice. Circulation 2006;114:1522–30
  • Smyth MJ, Takeda K, Hayakawa Y, et al. Nature's TRAIL–on a path to cancer immunotherapy. Immunity 2003;18:1–6
  • Watt V, Chamberlain J, Steiner T, et al. TRAIL attenuates the development of atherosclerosis in apolipoprotein E deficient mice. Atherosclerosis 2011;215:348–54

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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