Elsevier

Placenta

Volume 31, Issue 7, July 2010, Pages 561-567
Placenta

Current Topic
Dysregulated Complement Activation as a Common Pathway of Injury in Preeclampsia and Other Pregnancy Complications

https://doi.org/10.1016/j.placenta.2010.03.010Get rights and content

Abstract

The complement system protects the host against invading organisms, initiates inflammation and dispose of immune complexes and the products of inflammatory injury. The complement system provides an important link between the innate and adaptive immune systems. Experimental observations suggest that increased complement activation causes and/or perpetuates inflammation during pregnancy. Recent studies suggest a link between complement activation and preeclampsia. Excessive activation or insufficient regulation of complement recruits leukocytes and unleashes potent inflammatory and anti-angiogenic mediators associated with placental insufficiency and maternal endothelial dysfunction characteristic of preeclampsia. We review the animal and human studies that link complement activation and pathogenic events in preeclampsia, present evidence that activation of the complement system is associated with the development of preeclampsia and provides new targets to prevent its complications.

Section snippets

Angiogenic and immune dysregulation in preeclampsia

Preeclampsia is a complex multisystem disease that contributes significantly to maternal and neonatal mortality and morbidity [1], [2], [3]. The classical clinical manifestations, de novo hypertension and proteinuria, occur late in pregnancy, in the setting of maternal endothelial cell activation [4] and excessive systemic inflammation [5].

Preeclampsia has been called the “disease of theories” as its cause is unclear [2]. The pathologic process originates in the placenta, with inadequate

The complement system and tissue injury

The complement system, composed of over 30 proteins that act in concert to protect the host against invading organisms, initiates inflammation and tissue injury (Fig. 2) (described in [45], [46], [47]). Like the clotting cascade, the complement cascade contains a potent amplification mechanism based on sequential cleavage of inactive zymogen forms of proteins by serine protease mechanisms. Complement activation promotes chemotaxis of inflammatory cells and generates proteolytic fragments that

Models of the antiphospholipid antibody syndrome and other adverse pregnancy outcomes

That appropriate complement inhibition is an absolute requirement for normal pregnancy has been demonstrated by the finding that deficiency of Crry (a membrane-bound intrinsic complement regulatory protein in mice, like DAF and MCP, that blocks classical and alternative pathway activation of C3 [62]) leads to embryonic lethality in mice; Crry−/− embryos are surrounded by activated C3 fragments and PMN [63]. Importantly, embryos are rescued when C3 deficiency or factor B deficiency is introduced

Complement and human pregnancy

Little information is available about complement activation in normal and abnormal human pregnancy [73]. During normal gestation, serum levels of C3, C4, and total hemolytic complement (CH50) gradually increase 10–50% [74], likely reflecting increased synthesis, and levels of complement split products increase, suggesting low grade classical pathway activation [75]. Studies performed nearly 20 years ago showed marked elevations in levels of Bb, C3a, C4d, and soluble C5b-9 in preeclampsia,

Complement activation in preeclampsia patients

In 2005, inspired by novel research in the animal model of APS [65], [90], [91], Lynch et al conducted a prospective study in human pregnancy (n = 701) to investigate whether elevated levels of complement activation fragments Bb (reflecting alternative complement pathway activation) at a single point in early pregnancy (less than 20 weeks gestation) were predictive of preeclampsia later in pregnancy [92]. This was the first large prospective study to examine the relationship between Bb levels in

Conclusion

The complement system provides a link between the innate and adaptive immune systems, recognizing and responding to danger. In preeclampsia, perhaps related to immune maladaptation or oxidative stress, there is increased complement activation at the maternal–fetal interface. Excessive activation or insufficient regulation of complement recruits leukocytes and unleashes potent inflammatory and anti-angiogenic mediators associated with placental insufficiency and maternal endothelial dysfunction

Acknowledgements

This research was supported in part by NIH AR49772 (JES), AR38889 (JES) and K23 HD049684 (AML), the Mary Kirkland Center for Lupus Research (JES), American Heart Association (AML), the Center for Women’s Health Research and the List Family Foundation at University of Colorado Denver (AML) and Newborn Hope Colorado (AML).

References (104)

  • B. Giannakopoulos et al.

    How we diagnose the antiphospholipid syndrome

    Blood

    (2009)
  • S. Miyakis et al.

    International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS)

    J Thromb Haemost

    (2006)
  • G. Moroni et al.

    Pregnancy in lupus nephritis

    Am J Kidney Dis

    (2002)
  • J. Milliez et al.

    The prevalence of autoantibodies during third-trimester pregnancy complicated by hypertension or idiopathic fetal growth retardation

    Am J Obstet Gynecol

    (1991)
  • A.P. Sjoberg et al.

    Complement activation and inhibition: a delicate balance

    Trends Immunol

    (2009)
  • C.H. Holmes et al.

    Complement and pregnancy: new insights into the immunobiology of the fetomaternal relationship

    Baillieres Clin Obstet Gynaecol

    (1992)
  • B.P. Morgan et al.

    Immunology of reproduction: protecting the placenta

    Curr Biol

    (2000)
  • A.K. Abbas et al.

    Immunology: improving on nature in the twenty-first century

    Cell

    (2000)
  • M. Wells et al.

    Complement component deposition in uteroplacental (spiral) arteries in normal human pregnancy

    J Reprod Immunol

    (1987)
  • D.S. Cunningham et al.

    Decay-accelerating factor protects human trophoblast from complement-mediated attack

    Clin Immunol Immunopathol

    (1995)
  • P.F. Zipfel et al.

    FHL-1/reconectin: a human complement and immune regulator with cell-adhesive function

    Immunol Today

    (1999)
  • A.L. Stahl et al.

    Factor H dysfunction in patients with atypical hemolytic uremic syndrome contributes to complement deposition on platelets and their activation

    Blood

    (2008)
  • A. Richards et al.

    Inherited complement regulatory protein deficiency predisposes to human disease in acute injury and chronic inflammatory states the examples of vascular damage in atypical hemolytic uremic syndrome and debris accumulation in age-related macular degeneration

    Adv Immunol

    (2007)
  • D. Mao et al.

    Negligible role of antibodies and C5 in pregnancy loss associated exclusively with C3-dependent mechanisms through complement alternative pathway

    Immunity

    (2003)
  • P. Redecha et al.

    Tissue factor: a link between C5a and neutrophil activation in antiphospholipid antibody induced fetal injury

    Blood

    (2007)
  • M. Empson et al.

    Recurrent pregnancy loss with antiphospholipid antibody: a systematic review of therapeutic trials

    Obstet Gynecol

    (2002)
  • H.J. Imrie et al.

    Reduction in erythrocyte complement receptor 1 (CR1, CD35) and decay accelerating factor (DAF, CD55) during normal pregnancy

    J Reprod Immunol

    (1996)
  • R. Rampersad et al.

    The C5b–9 membrane attack complex of complement activation localizes to villous trophoblast injury in vivo and modulates human trophoblast function in vitro

    Placenta

    (2008)
  • J.M. Shamonki et al.

    Excessive complement activation is associated with placental injury in patients with antiphospholipid antibodies

    Am J Obstet Gynecol

    (2007)
  • J.P. Buyon et al.

    Serum complement values (C3 and C4) to differentiate between systemic lupus activity and pre-eclampsia

    Am J Med

    (1986)
  • F. Fakhouri et al.

    Factor H, membrane cofactor protein, and factor I mutations in patients with hemolysis, elevated liver enzymes, and low platelet count syndrome

    Blood

    (2008)
  • J.M. Thurman et al.

    A novel inhibitor of the alternative complement pathway prevents antiphospholipid antibody-induced pregnancy loss in mice

    Mol Immunol

    (2005)
  • A.M. Lynch et al.

    Alternative complement pathway activation fragment Bb in early pregnancy as a predictor of preeclampsia

    Am J Obstet Gynecol

    (2008)
  • A. Lynch et al.

    Early complement activation and subsequent risk for gestational hypertension

    Am J Obstet Gynecol

    (2008)
  • A.M. Lynch et al.

    Complement activation fragment Bb in early pregnancy and spontaneous preterm birth

    Am J Obstet Gynecol

    (2008)
  • J. Caprioli et al.

    Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome

    Blood

    (2006)
  • C.J. Fang et al.

    Membrane cofactor protein mutations in atypical hemolytic uremic syndrome (aHUS), fatal Stx-HUS, C3 glomerulonephritis, and the HELLP syndrome

    Blood

    (2008)
  • V. Fremeaux-Bacchi et al.

    Mutations in complement C3 predispose to development of atypical hemolytic uremic syndrome

    Blood

    (2008)
  • J. Zhang et al.

    Severe maternal morbidity associated with hypertensive disorders in pregnancy in the United States

    Hypertens Pregnancy

    (2003)
  • J.M. Roberts

    Endothelial dysfunction in preeclampsia

    Semin Reprod Endocrinol

    (1998)
  • T. Cindrova-Davies

    Gabor Than Award Lecture 2008: pre-eclampsia – from placental oxidative stress to maternal endothelial dysfunction

    Placenta

    (2009)
  • P. Parham

    NK cells and trophoblasts: partners in pregnancy

    J Exp Med

    (2004)
  • K. Red-Horse et al.

    Cytotrophoblast induction of arterial apoptosis and lymphangiogenesis in an in vivo model of human placentation

    J Clin Invest

    (2006)
  • G.J. Burton et al.

    Placental oxidative stress: from miscarriage to preeclampsia

    J Soc Gynecol Investig

    (2004)
  • G.J. Burton et al.

    Placental endoplasmic reticulum stress and oxidative stress in the pathophysiology of unexplained intrauterine growth restriction and early onset preeclampsia

    Placenta

    (2009)
  • G. Girardi et al.

    Complement activation induces dysregulation of angiogenic factors and causes fetal rejection and growth restriction

    J Exp Med

    (2006)
  • R.J. Levine et al.

    Circulating angiogenic factors and the risk of preeclampsia

    N Engl J Med

    (2004)
  • S.E. Maynard 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)
  • C.W. Redman et al.

    Placental stress and pre-eclampsia: a revised view

    Placenta

    (2009)
  • J.M. Roberts et al.

    The two stage model of preeclampsia: variations on the theme

    Placenta

    (2009)
  • Cited by (87)

    • Risk Factors for Preeclampsia

      2023, Hypertension: A Companion to Braunwald's Heart Disease
    • Diseases and conditions that impact maternal and fetal health and the potential for nanomedicine therapies

      2021, Advanced Drug Delivery Reviews
      Citation Excerpt :

      These mice spontaneously have fetal growth restriction, abnormal placentation, and defects in maternal decidual arteries [76,77]. These issues lead to inflammation and activation of the complement pathway, which has been shown to be associated with adverse pregnancy outcomes [78–80]. Complement inhibition using a fusion protein (CR2-Crry) between a pan-C3 convertase inhibitor, Crry, and a targeting protein that binds to C3 degradation products, CR2, was able to increase placental weight, normalize junctional zone and spiral artery morphology, increase placental vascular endothelial growth factor (VEGF) concentration, and reduce neutrophil recruitment to the placenta without altering the numbers of uterine natural killer cells or macrophages [81].

    • Genetic Factors in the Etiology of Preeclampsia/Eclampsia

      2021, Chesley’s Hypertensive Disorders in Pregnancy
    View all citing articles on Scopus
    View full text