Role of vascular endothelial growth factor and ...

1 downloads 0 Views 1MB Size Report
Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth factor ... Simmons LA, Hennessy A, Gillin AG et al. Uteroplacental blood flow and placental ...
doi:10.1111/jog.12751

J. Obstet. Gynaecol. Res. 2015

Role of vascular endothelial growth factor and placental growth factor expression on placenta structure in preeclamptic pregnancy Emel Kurtoglu1, Berrin Z. Altunkaynak2, Isinsu Aydin2, Ayse Z. Ozdemir1, Gamze Altun2, Arif Kokcu1 and Suleyman Kaplan2 Departments of 1Obstetrics and Gynecology, 2Histology and Embryology, Ondokuz Mayis University, Samsun, Turkey

Abstract Aim: Pre-eclampsia is a hypertensive disease that is characterized by high blood pressure and proteinuria after 20 gestational weeks and complicates 3–8% of all pregnancies. It is classified as either mild or severe pre-eclampsia according to severity, and the aim of this study was to investigate the structural differences between these two classifications. Methods: Placenta samples were collected from 68 women who underwent cesarean delivery. Total volume of villi and numerical density of vascular endothelial growth factor (VEGF)- and placental growth factor (PIGF)-positive cells were estimated on stereology and evaluated using one-way ANOVA. Results: There was no significantly difference in total villi volumes between the groups (P > 0.05). In contrast, on immunohistochemistry, the numerical density of VEGF-positive cells in severe pre-eclampsia was significantly different to the control and mild pre-eclampsia groups (P 0.05; Figs 5,7).

Discussion It has long been known that pregnancies complicated by pre-eclampsia have macroscopic and microscopic alterations in placental structure. In normal placentation, the cytotrophoblast cells invade the uterine spiral arteries and transform them from small-caliber resistance vessels into high-caliber capacitance vessels in order to

4

provide adequate perfusion for both villous and fetal growth.28,29 In pre-eclampsia, this invasion is limited to the proximal decidua, and spiral arteries fail to dilate and remain responsive to vasomotor influences, which leads to high-resistance chorioplacental circulation.30,31 As a result, the average diameter of the blood vessels decreases and, owing to the placental ischemia accelerating villous branching, causes an increase in total villi volume, and occurrence of numerous syncytial knots (apoptotic cell clusters) associated with diffusion and villitis.26,32–37 In contrast to the previous findings, there was no significant difference in mean volume of villi between the control and the pre-eclamptic groups. On the other hand, mean capillary volume in the villi was found to be increased in both mild and severe pre-eclamptic groups compared with the controls. Chronic hypoxia may have induced the growth of capillaries, increasing mean volume, but capillary loops with narrow lumens may have led to insufficient maternal–fetal perfusion. In addition, perivillous fibrin deposits and increased syncytial knot density owing to reduced perfusion and oxidative stress injury may have obstructed the hyperramification of the villi and decreased maternofetal exchange.

© 2015 Japan Society of Obstetrics and Gynecology

VEGF and PIGF in pre-eclamptic placenta

Figure 3 (a–c) H&E-stained placental sections in the (a) control group; (b) mild pre-eclampsia group; and (c) severe pre-eclampsia group. White arrows, syncytial knots; black arrows, perivillous fibrinoid deposition; black arrowheads, thrombosis areas. Although there was no significantly difference in terms of total villi volume between the groups (P > 0.05), thrombosis was noted in (b,c) the mild and severe pre-eclampsia groups on histopathology. Additionally, (a) perivillous fibrin deposition was observed in the control group. (Scale bars, 500 μm; insets, 250 μm).

Figure 4 Mean numerical density of placental growth factor- and vascular endothelial growth factorpositive cells per mm2 was estimated using unbiased counting frame for each section. X, Cells touching the red edges were excluded from the counting; √, cells that touched the green edges were included in the counting.

Figure 5 Numerical density of (a) placental growth factor- and (b) vascular endothelial growth factor-positive cells in the control and mild and severe pre-eclampsia groups. Error bars, SEM. *P < 0.05, **,##P < 0.01.

Given that defects in angiogenesis and remodeling of the trophoblast lead to maldevelopment, and inadequate placental mass, placental dysfunction, and chronic hypoxemia play key roles in the pathogenesis of preeclampsia, VEGF and PIGF and pro-angiogenic proteins produced by villous syncytiotrophoblasts have been the focus of many studies.17,18,38,39 Maternal serum and placenta levels of these pro-angiogenic factors have been

© 2015 Japan Society of Obstetrics and Gynecology

investigated in many studies, but there has been little interest in the immunohistochemistry of the expression of these factors in the placenta. Some of the previous studies comparing placental expression of the VEGF system, which is known to be regulated in response to mainly placental hypoxia, noted increased placental VEGF and PIGF immunostaining in pre-eclampsia pregnancies.14,17,18,40 Induction and

5

E. Kurtoglu et al.

Figure 6 Immunohistochemical localization of vascular endothelial growth factor (VEGF) in the (a) control group; (b) mild preeclampsia group; and (c) severe pre-eclampsia group. There are more VEGF-specific cells (white arrows) in the (c) severe preeclampsia group than in the other groups. Black stars, vessels; white arrows, syncytial knots. Scale bars, 25 μm.

Figure 7 Immunohistochemical localization of placental growth factor (PIGF) in the (a) control group; (b) mild pre-eclampsia group; and (c) severe pre-eclampsia group. The number of PIGF-specific cells (black arrows) in the (b) mild and (c) severe pre-eclampsia groups was significantly higher than in the (a) control group (P < 0.01). White stars, vessels; black arrowheads, syncytial knots. Scale bars, 25 μm.

upregulation of the VEGF system by chronic hypoxia and endothelial activation were suggested as possible mechanisms of this increase. In contrast, most of the studies reported decreased placental VEGF and PIGF immunostaining.12,13,38,41,42 In one of these investigations, VEGF was thought to be decreased due to insufficient compensation of the placenta.43 In addition, PIGF was thought to be lower in the placentas of pre-eclamptic patients due to reduced development of placental structures.44 Moreover, disregulation in the VEGF family and two possible mechanisms were suggested to explain the defective angiogenesis: one was downregulation of membrane-bound VEGF receptor-1 (VEGFR-1) in placentas leading to defective uteroplacental development; and the second was overproduction of competitive soluble VEGFR-1, which may lead to suppression of VEGF and PIGF effects.45 In the present study, VEGF was found to be significantly increased in the placentas of severe pre-eclamptic patients when compared with mild pre-eclampsia and the controls. It is possible that the hypoxic environment

6

that induces VEGF production has different features in terms of severity and enhancing factors in mild and severe cases, such that VEGF remained unchanged in mild cases. But, despite elevated levels, disregulation at the receptor level leading to dysfunction of VEGF and defective angiogenesis may have occurred. In this study, PIGF was also found to have increased in both mild and severe pre-eclampsia when compared with the controls, whereas there was no significant difference in PIGF between mild and severe pre-eclampsia. Despite the fact that this pro-angiogenic factor is induced by chronic hypoxia, the severity of hypoxia and oxidative injury and structural maldevelopment of the placenta may have affected the level of PIGF in the preeclampsia groups. Also, elevated levels seemed to be ineffective to supply adequate maternofetal circulation, which may have been caused by a defective function of the ligand–receptor complex of PIGF. Furthermore, in contrast to the previous studies, PIGF was found to be elevated in mild pre-eclamptic patients whose duration of pregnancy was 38.1±1.7 weeks and in severe pre-eclamptic

© 2015 Japan Society of Obstetrics and Gynecology

VEGF and PIGF in pre-eclamptic placenta

patients whose duration of pregnancy was 34.0±4.3 weeks. It may be that increase in PIGF does not depend on preterm delivery. In conclusion, placenta structure, particularly vascularization, reflects a complex interaction of regulatory factors including predominantly growth factors. Given that pre-eclampsia has been shown to have placentarelated vascular insufficiencies that lead to alterations in structure, understanding the features of the regulatory system will provide insight into the pathogenesis of preeclampsia.

Disclosure The authors report no conflict of interest. The authors alone are responsible for the content and writing of this paper.

References 1. American College of Obstetricians and Gynecologists, Task Force on Hypertension in Pregnancy. Hypertension in pregnancy: Report of the American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy. Obstet Gynecol 2013; 122: 1122–1131. 2. Roberts JM, Lain KY. Recent insights into the pathogenesis of pre-eclampsia. Placenta 2002; 23: 359–372. 3. Polliotti BM, Fry AG, Saller DN et al. Second-trimester maternal serum placental growth factor and vascular endothelial growth factor for predicting severe, early-onset preeclampsia. Obstet Gynecol 2003; 101: 1266–1274. 4. 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: 177–182. 5. Masuyama H, Suwaki N, Nakatsukasa H et al. Circulating angiogenic factors in preeclampsia, gestational proteinuria, and preeclampsia superimposed on chronic glomerulonephritis. Am J Obstet Gynecol 2006; 194: 551–556. 6. 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: 649–658. 7. Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci USA 1993; 90: 10705–10709. 8. Levine RJ, Maynard SE, Qian C. et al. Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med 2004; 350: 672–683. 9. Torry DS, Torry RJ. Angiogenesis and the expression of vascular endothelial growth factor in endometrium and placenta. Am J Reprod Immunol 1997; 37: 21–29. 10. Jackson MR, Carney EW, Lye SJ et al. Localization of two angiogenic growth factors (PDECGF and VEGF) in human placentae throughout gestation. Placenta 1994; 15: 341–353.

© 2015 Japan Society of Obstetrics and Gynecology

11. Shore VH, Wang TH, Wang CL et al. Vascular endothelial growth factor, placenta growth factor and their receptors in isolated human trophoblast. Placenta 1997; 18: 657–665. 12. Lyall F, Young A, Boswell F et al. Placental expression of vascular endothelial growth factor in placentae from pregnancies complicated by pre-eclampsia and intrauterine growth restriction does not support placental hypoxia at delivery. Placenta 1997; 18: 269–276. 13. Cheng Z, Lin Q, Shen Z. Study on association of vascular endothelial growth factor with the pathogenesis of pregnancy induced hypertension. Zhonghua Fu Chaan Ke Za Zhi 2001; 36: 72–75. 14. Simmons LA, Hennessy A, Gillin AG et al. Uteroplacental blood flow and placental vascular endothelial growth factor in normotensive and pre-eclamptic pregnancy. BJOG 2000; 107: 678–685. 15. Everet RB, Worlwy RC, Mc Donald PC et al. Effect of prostaglandin synthetase inhibitors on prosser responce to Angiotension II in human pregnancy. J Clin Endocrinol Metab 1978; 46: 1007–1010. 16. Redman CWG. Immunolgy of preeclampsia. Semin Perinatol 1991; 15: 257–262. 17. Dubova EA, Pavlov KA, Lyapin VM et al. Vascular endothelial growth factor and its receptors in the placental villi of pregnant patients with pre-eclampsia. Bull Exp Biol Med 2013; 154: 792–795. 18. Akercan F, Cirpan T, Terek MC et al. The immunohistochemical evaluation of VEGF in placenta biopsies of pregnancies complicated by preeclampsia. Arch Gynecol Obstet 2008; 277: 109–114. 19. Muy-Rivera M, Vadachkoria S, Woelk GB et al. Maternal Plasma VEGF, sVEGF-R1, and PlGF Concentrations in Preeclamptic and Normotensive Pregnant Zimbabwean. Women Physiol Res 2005; 54: 611–622. 20. Fisher SJ. The placental problem: linking abnormal cytotrophoblast differentiation to the maternal symptoms of preeclampsi. Reprod Biol Endocrinol 2004; 2: 53–56. 21. Zhou Y, Damsky CH, Chiu K et al. Preeclampsia is associated with abnormal expression of adhesion molecules by invasive cytotrophoblasts. J Clin Invest 1993; 91: 950–960. 22. Wathen KA, Tuutti E, Stenman UH et al. Maternal serum soluble vascular endothelial growth factor receptor 1 in early pregnancy ending in preeclampsia or intrauterine growth retardation. J Clin Endocrinol Metab 2006; 91: 180–184. 23. Maglione D, Guerriero V, Viglietto G et al. Isolation of a human placenta cDNA coding for a protein related to the vascular permeability factor. Proc Natl Acad Sci USA 1991; 88: 9267–9271. 24. Sahin B, Elfaki A. Estimation of the Volume and Volume Fraction of Brain and Brain Structures on Radiological Image. NeuroQuantology 2012; 10: 87–97. 25. Piskin A, Altunkaynak BZ, Tumen Temur G et al. The beneficial effects of Momordica charantia (bitter gourd) on wound healing of rabbit skin. Journal of Dermatological Treatment 2014; 25: 350–357. 26. Sankar KD, Bhanu PS, Kiran S et al. Vasculosyncytial membrane in relation to syncytial knots complicates the placenta in preeclampsia: a histomorphometrical study. Anat Cell Biol 2012; 45: 86–91. 27. Gundersen HJ, Jensen EB. The efficiency of systematic sampling in stereology and its prediction. J Microsc 1987; 147: 229–263. 28. Kaufmann P, Burton G. Anatomy and genesis of placenta. In: Knobil E, Neill JD(eds). The Physiology of Reproduction. New York: Raven Press, 1994; 441–483.

7

E. Kurtoglu et al.

29. Mayhew TM. Villous trophoblast of human placenta: a coherent view of its turnover, repair and contributions to villous development and maturation. Histol Histopathol 2001; 16: 1213–1224. 30. Saeed I, Iqbal I, Sarfaraz R et al. Histomorphological changes in placentae of preeclamptic mothers with reference to vasculosyncytial membrane thickness and syncytial knot formation. J Rawalpindi Med Coll 2012; 16: 51–54. 31. Pijnenborg R, Vercruysse L, Hanssens M. The uterine spiral arteries in human pregnancy: facts and controversies. Placenta 2006; 27: 939–958. 32. Peng M, Yu L, Ding YL, Zhou CJ. Trophoblast cells invaing the placenta bed and change of spiral arteries and microvessels in pre-eclampsia. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2008; 33: 121–129. 33. Burton GJ, Woods AW, Jauniaux E, Kingdom JC. Rheological and physiological consequences of conversion of the maternal spiral arteries for uteroplacental blood flow during human pregnancy. Placenta 2009; 30: 473–482. 34. Roberts DJ, Post MD. Post. J Clin Pathol 2008; 61: 1254–1260. 35. Kingdom J, Huppertz B, Seaward G et al. Development of the placental villous tree and its consequences for fetal growth. Eur J Obstet Gynecol Reprod Biol 2000; 92: 35–43. 36. Heazell AE, Moll SJ, Jones CJ et al. Formation of syncytial knots is increased by hyperoxia, hypoxia and reactive oxygen species. Placenta 2007; 28: 33–40. 37. Ducray JF, Naicker T, Moodley J. Pilot study of comparative placental morphometry in pre-eclamptic and normotensive pregnancies suggests possible maladaptations of the fetal component of the placenta. Eur J Obstet Gynecol Reprod Biol 2011; 156: 29–34.

8

38. Sezer SD, Kucuk M, Doger FK et al. VEGF, PIGF and HIF-1α in placentas of early- and late-onset pre-eclamptic patients. Gynecol Endocrinol 2013; 29: 797–800. 39. Ferrara N, Keyt B. Vascular endothelial growth factor: Basic biology and clinical implications. EXS 1997; 79: 209–232. 40. Trollmann R, Amann K, Schoof E et al. Hypoxia activates the human placental vascular endothelial growth factor system in vitro and in vivo: Up-regulation of vascular endothelial growth factor in clinically relevant hypoxic ischemia in birth asphyxia. Am J Obstet Gynecol 2003; 188: 517–523. 41. Sun SG, Shen N, Zheng YH et al. Expression of hypoxia-inducible factor-1alpha, vascular endothelial growth factor and sFlt-1 in preeclampsia placenta. Zhonghua Fu Chan Ke Za Zhi 2006; 41: 440–444. 42. Wang F, Bai H, Fan P et al. Vasohibin and VEGF protein levels in placentae from pregnancies complicated by severe pre-eclampsia]. Sichuan Da Xue Xue Bao Yi Xue Ban 2010; 41: 814–817. 43. Sgambati E, Marini M, Zappoli Thyrion GD et al. VEGF expression in the placenta from pregnancies complicated by hypertensive disorders. BJOG 2004; 111: 564–570. 44. 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: 177–182. 45. 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: 5555–5563.

© 2015 Japan Society of Obstetrics and Gynecology