Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Soluble endoglin contributes to the pathogenesis of preeclampsia

A Corrigendum to this article was published on 01 July 2006

Abstract

Preeclampsia is a pregnancy-specific hypertensive syndrome that causes substantial maternal and fetal morbidity and mortality. Maternal endothelial dysfunction mediated by excess placenta-derived soluble VEGF receptor 1 (sVEGFR1 or sFlt1) is emerging as a prominent component in disease pathogenesis. We report a novel placenta-derived soluble TGF-β coreceptor, endoglin (sEng), which is elevated in the sera of preeclamptic individuals, correlates with disease severity and falls after delivery. sEng inhibits formation of capillary tubes in vitro and induces vascular permeability and hypertension in vivo. Its effects in pregnant rats are amplified by coadministration of sFlt1, leading to severe preeclampsia including the HELLP (hemolysis, elevated liver enzymes, low platelets) syndrome and restriction of fetal growth. sEng impairs binding of TGF-β1 to its receptors and downstream signaling including effects on activation of eNOS and vasodilation, suggesting that sEng leads to dysregulated TGF-β signaling in the vasculature. Our results suggest that sEng may act in concert with sFlt1 to induce severe preeclampsia.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Expression of ENG mRNA and Eng in placentae of normal and preeclamptic pregnancies.
Figure 2: Increased sEng levels in sera from individuals with preeclampsia.
Figure 3: sEng inhibits capillary formation and increases vascular permeability.
Figure 4: Renal, placental and hepatic histological changes and peripheral blood smears in pregnant rats after sEng and sFlt1 treatment.
Figure 5: Recombinant sEng attenuates TGF-β1 binding and has effects on vasodilation through activation of eNOS.

Similar content being viewed by others

Evdokia Dimitriadis, Daniel L. Rolnik, … Ellen Menkhorst

Notes

  1. NOTE: Due to an editing error, some reference citations in the text are incorrect. In particular, the second citation of ref. 22 (on p. 644) should be ref. 23 and refs. 23–49 should be refs. 24–50. This has been corrected in the HTML and PDF versions of the article.

References

  1. Sibai, B., Dekker, G. & Kupferminc, M. Pre-eclampsia. Lancet 365, 785–799 (2005).

    Article  Google Scholar 

  2. Weinstein, L. Syndrome of hemolysis, elevated liver enzymes, and low platelet count: a severe consequence of hypertension in pregnancy. Am. J. Obstet. Gynecol. 142, 159–167 (1982).

    Article  CAS  Google Scholar 

  3. Roberts, J.M. et al. Preeclampsia: an endothelial cell disorder. Am. J. Obstet. Gynecol. 161, 1200–1204 (1989).

    Article  CAS  Google Scholar 

  4. Maynard, S.E. et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J. Clin. Invest. 111, 649–658 (2003).

    Article  CAS  Google Scholar 

  5. Zhou, Y. et al. Vascular endothelial growth factor ligands and receptors that regulate human cytotrophoblast survival are dysregulated in severe preeclampsia and hemolysis, elevated liver enzymes, and low platelets syndrome. Am. J. Pathol. 160, 1405–1423 (2002).

    Article  CAS  Google Scholar 

  6. Ahmad, S. & Ahmed, A. Elevated placental soluble vascular endothelial growth factor receptor-1 inhibits angiogenesis in preeclampsia. Circ. Res. 95, 884–891 (2004).

    Article  CAS  Google Scholar 

  7. Chaiworapongsa, T. 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. 190, 1541–1547; discussion 1547–1550 (2004).

    Article  CAS  Google Scholar 

  8. Taylor, R.N. et al. Longitudinal serum concentrations of placental growth factor: evidence for abnormal placental angiogenesis in pathologic pregnancies. Am. J. Obstet. Gynecol. 188, 177–182 (2003).

    Article  CAS  Google Scholar 

  9. Levine, R.J. et al. Circulating angiogenic factors and the risk of preeclampsia. N. Engl. J. Med. 350, 672–683 (2004).

    Article  CAS  Google Scholar 

  10. Chaiworapongsa, T. 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. 17, 3–18 (2005).

    Article  CAS  Google Scholar 

  11. Hertig, A. et al. Maternal serum sFlt1 concentration is an early and reliable predictive marker of preeclampsia. Clin. Chem. 50, 1702–1703 (2004).

    Article  CAS  Google Scholar 

  12. Romero, R. et al. Clinical significance, prevalence, and natural history of thrombocytopenia in pregnancy-induced hypertension. Am. J. Perinatol. 6, 32–38 (1989).

    Article  CAS  Google Scholar 

  13. Cheifetz, S. et al. Endoglin is a component of the transforming growth factor-beta receptor system in human endothelial cells. J. Biol. Chem. 267, 19027–19030 (1992).

    CAS  PubMed  Google Scholar 

  14. Gougos, A. et al. Identification of distinct epitopes of endoglin, an RGD-containing glycoprotein of endothelial cells, leukemic cells, and syncytiotrophoblasts. Int. Immunol. 4, 83–92 (1992).

    Article  CAS  Google Scholar 

  15. St-Jacques, S., Forte, M., Lye, S.J. & Letarte, M. Localization of endoglin, a transforming growth factor-beta binding protein, and of CD44 and integrins in placenta during the first trimester of pregnancy. Biol. Reprod. 51, 405–413 (1994).

    Article  CAS  Google Scholar 

  16. McAllister, K.A. et al. Endoglin, a TGF-β binding protein of endothelial cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat. Genet. 8, 345–351 (1994).

    Article  CAS  Google Scholar 

  17. Bourdeau, A., Dumont, D.J. & Letarte, M. A murine model of hereditary hemorrhagic telangiectasia. J. Clin. Invest. 104, 1343–1351 (1999).

    Article  CAS  Google Scholar 

  18. Li, D.Y. et al. Defective angiogenesis in mice lacking endoglin. Science 284, 1534–1537 (1999).

    Article  CAS  Google Scholar 

  19. Toporsian, M. et al. A role for endoglin in coupling eNOS activity and regulating vascular tone revealed in hereditary hemorrhagic telangiectasia. Circ. Res. 96, 684–692 (2005).

    Article  CAS  Google Scholar 

  20. Dimmeler, S., Dernbach, E. & Zeiher, A.M. Phosphorylation of the endothelial nitric oxide synthase at ser-1177 is required for VEGF-induced endothelial cell migration. FEBS Lett. 477, 258–262 (2000).

    Article  CAS  Google Scholar 

  21. Garcia-Cardena, G. et al. Dynamic activation of endothelial nitric oxide synthase by Hsp90. Nature 392, 821–824 (1998).

    Article  CAS  Google Scholar 

  22. Fleming, I., Fisslthaler, B., Dimmeler, S., Kemp, B.E. & Busse, R. Phosphorylation of Thr(495) regulates Ca(2+)/calmodulin-dependent endothelial nitric oxide synthase activity. Circ. Res. 88, E68–E75 (2001).

    CAS  PubMed  Google Scholar 

  23. Brown, M.A., Zammit, V.C. & Lowe, S.A. Capillary permeability and extracellular fluid volumes in pregnancy-induced hypertension. Clin. Sci. (Lond.) 77, 599–604 (1989).

    Article  CAS  Google Scholar 

  24. Inoue, N. et al. Molecular regulation of the bovine endothelial cell nitric oxide synthase by transforming growth factor-beta 1. Arterioscler. Thromb. Vasc. Biol. 15, 1255–1261 (1995).

    Article  CAS  Google Scholar 

  25. Saura, M. et al. Smad2 mediates transforming growth factor-β induction of endothelial nitric oxide synthase expression. Circ. Res. 91, 806–813 (2002).

    Article  CAS  Google Scholar 

  26. Bdolah, Y., Sukhatme, V.P. & Karumanchi, S.A. Angiogenic imbalance in the pathophysiology of preeclampsia: newer insights. Semin. Nephrol. 24, 548–556 (2004).

    Article  Google Scholar 

  27. Redman, C.W. & Sargent, I.L. Latest advances in understanding preeclampsia. Science 308, 1592–1594 (2005).

    Article  CAS  Google Scholar 

  28. Li, C. et al. Plasma levels of soluble CD105 correlate with metastasis in patients with breast cancer. Int. J. Cancer 89, 122–126 (2000).

    Article  CAS  Google Scholar 

  29. Velasco-Loyden, G., Arribas, J. & Lopez-Casillas, F. The shedding of betaglycan is regulated by pervanadate and mediated by membrane type matrix metalloprotease-1. J. Biol. Chem. 279, 7721–7733 (2004).

    Article  CAS  Google Scholar 

  30. Benian, A., Madazli, R., Aksu, F., Uzun, H. & Aydin, S. Plasma and placental levels of interleukin-10, transforming growth factor-β1, and epithelial-cadherin in preeclampsia. Obstet. Gynecol. 100, 327–331 (2002).

    CAS  PubMed  Google Scholar 

  31. Muy-Rivera, M. et al. Transforming growth factor-β1 (TGF-β1) in plasma is associated with preeclampsia risk in Peruvian women with systemic inflammation. Am. J. Hypertens. 17, 334–338 (2004).

    Article  CAS  Google Scholar 

  32. Hennessy, A. et al. Transforming growth factor-β 1 does not relate to hypertension in pre-eclampsia. Clin. Exp. Pharmacol. Physiol. 29, 968–971 (2002).

    Article  CAS  Google Scholar 

  33. Barbara, N.P., Wrana, J.L. & Letarte, M. Endoglin is an accessory protein that interacts with the signaling receptor complex of multiple members of the transforming growth factor-beta superfamily. J. Biol. Chem. 274, 584–594 (1999).

    Article  CAS  Google Scholar 

  34. Shesely, E.G. et al. Elevated blood pressures in mice lacking endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 93, 13176–13181 (1996).

    Article  CAS  Google Scholar 

  35. Lowe, D.T. Nitric oxide dysfunction in the pathophysiology of preeclampsia. Nitric Oxide 4, 441–458 (2000).

    Article  CAS  Google Scholar 

  36. Papapetropoulos, A., Garcia-Cardena, G., Madri, J.A. & Sessa, W.C. Nitric oxide production contributes to the angiogenic properties of vascular endothelial growth factor in human endothelial cells. J. Clin. Invest. 100, 3131–3139 (1997).

    Article  CAS  Google Scholar 

  37. Predescu, D., Predescu, S., Shimizu, J., Miyawaki-Shimizu, K. & Malik, A.B. Constitutive eNOS-derived nitric oxide is a determinant of endothelial junctional integrity. Am. J. Physiol. Lung Cell. Mol. Physiol. 289, L371–L381 (2005).

    Article  CAS  Google Scholar 

  38. Tatsumi, M., Kishi, Y., Miyata, T. & Numano, F. Transforming growth factor-beta(1) restores antiplatelet function of endothelial cells exposed to anoxia-reoxygenation injury. Thromb. Res. 98, 451–459 (2000).

    Article  CAS  Google Scholar 

  39. Ristimaki, A., Ylikorkala, O. & Viinikka, L. Effect of growth factors on human vascular endothelial cell prostacyclin production. Arteriosclerosis 10, 653–657 (1990).

    Article  CAS  Google Scholar 

  40. He, H. et al. Vascular endothelial growth factor signals endothelial cell production of nitric oxide and prostacyclin through flk-1/KDR activation of c-Src. J. Biol. Chem. 274, 25130–25135 (1999).

    Article  CAS  Google Scholar 

  41. Mills, J.L. et al. Prostacyclin and thromboxane changes predating clinical onset of preeclampsia: a multicenter prospective study. J. Am. Med. Assoc. 282, 356–362 (1999).

    Article  CAS  Google Scholar 

  42. Darland, D.C. et al. Pericyte production of cell-associated VEGF is differentiation-dependent and is associated with endothelial survival. Dev. Biol. 264, 275–288 (2003).

    Article  CAS  Google Scholar 

  43. Zhou, Y. et al. Human cytotrophoblasts adopt a vascular phenotype as they differentiate. A strategy for successful endovascular invasion? J. Clin. Invest. 99, 2139–2151 (1997).

    Article  CAS  Google Scholar 

  44. Zhou, Y., Damsky, C.H. & Fisher, S.J. Preeclampsia is associated with failure of human cytotrophoblasts to mimic a vascular adhesion phenotype. One cause of defective endovascular invasion in this syndrome? J. Clin. Invest. 99, 2152–2164 (1997).

    Article  CAS  Google Scholar 

  45. Caniggia, I., Taylor, C.V., Ritchie, J.W., Lye, S.J. & Letarte, M. Endoglin regulates trophoblast differentiation along the invasive pathway in human placental villous explants. Endocrinology 138, 4977–4988 (1997).

    Article  CAS  Google Scholar 

  46. Fonsatti, E., Altomonte, M., Arslan, P. & Maio, M. Endoglin (CD105): a target for anti-angiogenetic cancer therapy. Curr. Drug Targets 4, 291–296 (2003).

    Article  CAS  Google Scholar 

  47. Holash, J. et al. VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc. Natl. Acad. Sci. USA 99, 11393–11398 (2002).

    Article  CAS  Google Scholar 

  48. ACOG practice bulletin. Diagnosis and management of preeclampsia and eclampsia. Number 33, January 2002. Obstet. Gynecol. 99, 159–167 (2002).

  49. Kuo, C.J. et al. Comparative evaluation of the antitumor activity of antiangiogenic proteins delivered by gene transfer. Proc. Natl. Acad. Sci. USA 98, 4605–4610 (2001).

    Article  CAS  Google Scholar 

  50. Akiyoshi, S. et al. c-Ski acts as a transcriptional co-repressor in transforming growth factor-beta signaling through interaction with smads. J. Biol. Chem. 274, 35269–35277 (1999).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all the staff in the Department of Obstetrics at the Beth Israel Deaconess Medical Center for help with patient identification and recruitment. We thank B. Furie's laboratory for help with measurement of platelet counts in rats, J. Min and the Physiology Core laboratory for help with blood pressure measurements, J. Li for vascular reactivity experiments, R. Mulligan for help with the production of sFlt1 adenoviruses, L. Zhang for technical assistance with mass spectrometry, B. Sachs, R. Levine, R. Thadhani, J. Flier, W. Aird and S. Pennathur for discussions. This work was funded by US National Institutes of Health grants DK064255 and HL079594 to S.A.K., Department of Medicine, Obstetrics and Gynecology seed funds to S.A.K. and V.P.S., and Heart and Stroke Foundation of Ontario grant T5016 to M.L.

Author information

Authors and Affiliations

Authors

Contributions

S.V.: mRNA and protein expression studies of Eng/sEng, generation and expression of sEng adenoviruses, all animal studies; substantial contribution to writing, editing and generation of figures. M.T.: biochemical characterization of sEng, TGF-β effects on eNOS dephosphorylation, studies of sEng on TGF-β effects; substantial contribution to writing and editing of manuscript and generation of figures. C.L.: enrollment and collection of all human material, ELISA studies on human samples, in vitro angiogenesis assays and biochemical studies of urine and plasma obtained from animals. J.H.: TGF-β–Smad promoter studies. T.M.: vascular permeability studies. Y.M.K.: immunohistochemistry of human placental samples. Y.B.: animal studies. K.H.L.: enrollment and collection of all human material, expertise in clinical preeclampsia. H.Y.: in vitro angiogenesis assays. T.A.L.: Affymetrix microarray experiments. I.E.S.: all rat histological studies, including electron microscopy. D.R.: rat placental histological studies. P.A.D.: provided expertise in vascular biology and TGF-β signaling. F.H.E.: provided expertise in clinical preeclampsia. F.W.S.: all microvascular reactivity experiments. R.R.: immunohistochemistry of human placental samples; provided expertise in preeclampsia. V.P.S.: interpretation of microarray experiments, microvascular permeability studies; provided overall expertise in vascular biology and preeclampsia; final editing of the manuscript. M.L.: provided expertise on Eng, analysis and interpretation of studies involving biochemical characterization of sEng, TGF-β effects on eNOS dephosphorylation, studies of sEng on TGF-β effects; substantially contributed to writing and editing of manuscript. S.A.K: principal investigator of the study; overall design and concept, analysis and interpretation of all data, drafting and final editing of the manuscript.

Corresponding author

Correspondence to S Ananth Karumanchi.

Ethics declarations

Competing interests

S. Ananth Karumanchi and Vikas P. Sukhatme are listed as co-inventors on multiple patents filed by the Beth Israel Deaconness Medical Center for the diagnosis and therapy of preeclampsia. These patents have been licensed to multiple diagnostic and therapeutic companies.

S. Ananth Karumanchi and Vikas P. Sukhatme serve as consultants to Abbott, Beckman Coulter and Johnson & Johnson.

Supplementary information

Supplementary Fig. 1

Electron microscopy documents glomerular endotheliosis in pregnant rats. (PDF 330 kb)

Supplementary Fig. 2

sEng inhibits TGF-β1–mediated vascular reactivity in mesenteric vessels. (PDF 208 kb)

Supplementary Fig. 3

Western blots of rat plasma demonstrating expression of the recombinant sFlt1 and sEng. (PDF 572 kb)

Supplementary Table 1

Clinical characteristics in the pregnant patient groups. (PDF 91 kb)

Supplementary Methods (PDF 88 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Venkatesha, S., Toporsian, M., Lam, C. et al. Soluble endoglin contributes to the pathogenesis of preeclampsia. Nat Med 12, 642–649 (2006). https://doi.org/10.1038/nm1429

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm1429

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing