Elsevier

Immunobiology

Volume 217, Issue 2, February 2012, Pages 195-203
Immunobiology

Review
Complement-mediated injury and protection of endothelium: Lessons from atypical haemolytic uraemic syndrome

https://doi.org/10.1016/j.imbio.2011.07.028Get rights and content

Abstract

The complement system provides a vital defence against invading pathogens. As an intrinsic system it is always ‘on’, in a state of constant, low level activation. This activation is principally mediated through the deposition of C3b on to pathogenic surfaces and host tissues. C3b is generated by spontaneous ‘tick over’ and formal activation of the alternative pathway, and by activation of the classical and lectin pathways. If the deposited C3b is not appropriately regulated, there is progression to terminal pathway complement activation via the C5 convertases, generating the potent anaphylotoxin C5a and the membrane attack complex C5b-9. Unsurprisingly, these highly active components have the potential to cause injury to bystander host tissue, including the vascular endothelium. As such, complement activation on endothelium is normally tightly controlled by a large number of fluid-phase and membrane bound inhibitors, in an attempt to ensure that propagation of complement activation is appropriately restricted to invading pathogens and altered ‘self’, e.g. apoptotic and necrotic cells.

The kidney is increasingly recognised as a site at particular risk from complement-mediated endothelial injury. Both genetic and acquired defects which impact on complement regulation predispose to this susceptibility. The thrombotic microangiopathy, haemolytic uraemic syndrome (HUS), will be used to illustrate the mechanisms by which the endothelial cell injury occurs. Finally, the underlying rationale for current and future potential therapeutic interventions in HUS and also the opportunities for enhancing endothelial defence to prevent relapsing disease through increased complement cytoprotective strategies will be summarised.

Section snippets

The endothelium

Endothelial cells line the interior surface of all blood vessels in the body, occupying a strategic location between the blood vessel walls and the blood stream. The endothelium is able to detect and respond to mechanical stimuli, such as pressure and shear stress, and to hormonal stimuli such as vasoactive mediators. Endothelial cells have important roles in health and disease. Endothelial cell functions include the release of agents which regulate vasomotor function, trigger inflammatory

Haemolytic uraemic syndrome (HUS)

HUS is a type of thrombotic microangiopathy and is characterised by the formation of fibrin-platelet clots in arterial microcirculations. There is an apparent predilection for the renal glomerular capillaries and arterioles, resulting in kidney failure, although the simultaneous involvement of other microvascular beds in the heart, brain and pancreas to a greater or lesser extent, is recognised and reported in all subtypes of HUS. Endothelial cell injury (swelling, detachment, and

The complement cascade

A principal biological function of the complement system is the rapid recognition and elimination of pathogens. Key mechanisms include opsonisation by C3b, activation of the inflammatory cascade by the generation of the anaphylotoxins C3a and C5a and lysis by the membrane attack complex (MAC; C5b-9). The complement cascade is activated by the classical pathway (CP) (Kojouharova et al. 2010), the alternative pathway (Harboe and Mollnes, 2008, Wallis et al., 2010) or the MBL/MASP pathway (Wallis

Endothelial protection against complement-mediated injury

The complement system is tightly controlled by natural fluid phase and membrane bound inhibitors, to restrict propagation to invading pathogens or altered self cells.

Endothelial cell activation and injury in HUS

Specific insights into the pivotal role of endothelial activation and injury as a trigger and a determinant of outcome in HUS will aid in understanding pathogenic mechanisms. Unravelling the links between genetic predisposition, inflammatory triggers, complement and the coagulation cascade will potentially identify therapeutic pathways with wider relevance for other conditions in which thrombotic vascular injury is a key feature, for example, ischaemia–reperfusion injury (IRI), preeclampsia and

Complement inhibition in aHUS

Current treatment protocols for aHUS are based on plasma exchange/infusion. The rationale for this treatment, which has significantly improved morbidity and mortality in individual cases, but has not been formally evaluated in a clinical trial, is logical. It may beneficially supplement levels of deficient complement components contained in plasma, e.g. Factor H or Factor I, secondly, it removes mutated proteins which may exert a dominant negative effect on complement regulation and thirdly it

Conclusions

Consideration of the interactions between complement and the endothelium is critical for understanding the pathophysiology of conditions with prominent endothelial cell injury. Unanswered questions in aHUS relate to reasons for the renal endothelial susceptibility; the determinants of penetrance in those with a genetic predisposition and the balance between soluble and membrane bound complement regulation at endothelial and basement membrane surfaces. Many aspects of endothelial cell biology

Acknowledgements

The authors gratefully acknowledge the support of their funding bodies. HK is a Medical Research Council Clinical Training Fellow and AR is a Wellcome Trust Intermediate Clinical Fellow. We would also like to thank David Kavanagh, Anne Astier and Jeremy Hughes for their careful consideration and valuable comments on the manuscript.

References (85)

  • D.E. Jenne et al.

    Clusterin: the intriguing guises of a widely expressed glycoprotein

    Trends Biochem. Sci.

    (1992)
  • A.R. Kinderlerer et al.

    KLF2-dependent, shear stress-induced expression of CD59: a novel cytoprotective mechanism against complement-mediated injury in the vasculature

    J. Biol. Chem.

    (2008)
  • A.R. Kinderlerer et al.

    Heme oxygenase-1 expression enhances vascular endothelial resistance to complement-mediated injury through induction of decay-accelerating factor: a role for increased bilirubin and ferritin

    Blood

    (2009)
  • M. Kojouharova et al.

    New insights into the molecular mechanisms of classical complement activation

    Mol. Immunol.

    (2010)
  • M.K. Liszewski et al.

    Inhibiting complement activation on cells at the step of C3 cleavage

    Vaccine

    (2008)
  • J.C. Lofling et al.

    A dietary non-human sialic acid may facilitate hemolytic-uremic syndrome

    Kidney Int.

    (2009)
  • J.A. Marcum et al.

    Cloned bovine aortic endothelial cells synthesize anticoagulantly active heparan sulfate proteoglycan

    J. Biol. Chem.

    (1986)
  • J.C. Mason et al.

    Decay-accelerating factor induction on vascular endothelium by vascular endothelial growth factor (VEGF) is mediated via a VEGF receptor-2 (VEGF-R2)- and protein kinase C-alpha/epsilon (PKCalpha/epsilon)-dependent cytoprotective signaling pathway and is inhibited by cyclosporin A

    J. Biol. Chem.

    (2004)
  • J.C. Mason et al.

    Induction of decay-accelerating factor by cytokines or the membrane-attack complex protects vascular endothelial cells against complement deposition

    Blood

    (1999)
  • A. Richards et al.

    Factor H mutations in hemolytic uremic syndrome cluster in exons 18–20, a domain important for host cell recognition

    Am. J. Hum. Genet.

    (2001)
  • A. Richards et al.

    Implications of the initial mutations in membrane cofactor protein (MCP; CD46) leading to atypical hemolytic uremic syndrome

    Mol. Immunol.

    (2007)
  • L.T. Roumenina et al.

    Hyperfunctional C3 convertase leads to complement deposition on endothelial cells and contributes to atypical hemolytic uremic syndrome

    Blood

    (2009)
  • C.Q. Schmidt et al.

    Production of biologically active complement factor H in therapeutically useful quantities

    Protein Expr. Purif.

    (2011)
  • N.S. Sheerin et al.

    TNF-alpha regulation of C3 gene expression and protein biosynthesis in rat glomerular endothelial cells

    Kidney Int.

    (1997)
  • P.J. Sims et al.

    Induction of cellular procoagulant activity by the membrane attack complex of complement

    Semin. Cell Biol.

    (1995)
  • R.A. Skidgel et al.

    Structure and function of human plasma carboxypeptidase N, the anaphylatoxin inactivator

    Int. Immunopharmacol.

    (2007)
  • A.L. Stahl et al.

    A novel mutation in the complement regulator clusterin in recurrent hemolytic uremic syndrome

    Mol. Immunol.

    (2009)
  • R. Wallis et al.

    Paths reunited: initiation of the classical and lectin pathways of complement activation

    Immunobiology

    (2010)
  • T.K. Blackmore et al.

    Identification of the second heparin-binding domain in human complement factor H

    J. Immunol.

    (1998)
  • A.M. Blom et al.

    A novel non-synonymous polymorphism (p.Arg240His) in C4b-binding protein is associated with atypical hemolytic uremic syndrome and leads to impaired alternative pathway cofactor activity

    J. Immunol.

    (2008)
  • L.N. Broekhuizen et al.

    Effect of sulodexide on endothelial glycocalyx and vascular permeability in patients with type 2 diabetes mellitus

    Diabetologia

    (2010)
  • R.A. Brooimans et al.

    Biosynthesis of complement factor H by human umbilical vein endothelial cells. Regulation by T cell growth factor and IFN-gamma

    J. Immunol.

    (1989)
  • R.A. Brooimans et al.

    CD59 expressed by human endothelial cells functions as a protective molecule against complement-mediated lysis

    Eur. J. Immunol.

    (1992)
  • R.A. Brooimans et al.

    Relative roles of decay-accelerating factor, membrane cofactor protein, and CD59 in the protection of human endothelial cells against complement-mediated lysis

    Eur. J. Immunol.

    (1992)
  • L.C. Clement et al.

    Podocyte-secreted angiopoietin-like-4 mediates proteinuria in glucocorticoid-sensitive nephrotic syndrome

    Nat. Med.

    (2011)
  • L. Copelovitch et al.

    Streptococcus pneumoniae-associated hemolytic uremic syndrome

    Pediatr. Nephrol.

    (2008)
  • H. Dauchel et al.

    Expression of complement alternative pathway proteins by endothelial cells. Differential regulation by interleukin 1 and glucocorticoids

    Eur. J. Immunol.

    (1990)
  • A. Davies et al.

    CD59, an LY-6-like protein expressed in human lymphoid cells, regulates the action of the complement membrane attack complex on homologous cells

    J. Exp. Med.

    (1989)
  • E.G. de Jorge et al.

    The development of atypical hemolytic uremic syndrome depends on complement C5

    J. Am. Soc. Nephrol.

    (2011)
  • M. Delvaeye et al.

    Thrombomodulin mutations in atypical hemolytic-uremic syndrome

    N. Engl. J. Med.

    (2009)
  • D.H. Endemann et al.

    Endothelial dysfunction

    J. Am. Soc. Nephrol.

    (2004)
  • V. Eremina et al.

    VEGF inhibition and renal thrombotic microangiopathy

    N. Engl. J. Med.

    (2008)
  • Cited by (74)

    • Targeting vascular inflammation through emerging methods and drug carriers

      2022, Advanced Drug Delivery Reviews
      Citation Excerpt :

      Endothelial cells tightly regulate cellular and humoral mechanisms of both innate and adaptive immune defense. The membrane-bound complement regulatory proteins, membrane cofactor protein and CD59 are constitutively expressed on healthy endothelial cells, to prevent undesired activation of complement and provide protection against complement-mediated injury [12]. The endothelium is also a key player in maintaining homeostasis of IgG, through the salvage pathway mediated by the neonatal Fc receptor (FcRn) [13–15].

    • The endocannabinoidome in neuropsychiatry: Opportunities and potential risks

      2021, Pharmacological Research
      Citation Excerpt :

      Several authors investigating the anti-inflammatory effects AEA on microglia in vitro have reported that such effects appear to be mediated by the activation of CB2R and subsequent downregulation of MAPK and NF-κB [521–524]. These findings have been broadly reproduced in studies examining the effects of AEA upregulation in vivo [525–529]. 2-AG also appears to ameliorate neuroinflammation by inhibiting NF-κB production in microglia and subsequent downregulation of PIC production [530–532].

    View all citing articles on Scopus
    View full text