Targeting pathogenic postischemic self-recognition by natural IgM to protect against posttransplantation cardiac reperfusion injury

Circulation. 2015 Mar 31;131(13):1171-80. doi: 10.1161/CIRCULATIONAHA.114.010482. Epub 2015 Feb 17.

Abstract

Background: Natural IgM antibodies represent a class of innate pattern recognition receptors that recognize danger-associated molecular patterns expressed on stressed or dying cells. They play important roles in tissue homeostasis by disposing of prenecrotic cells and suppressing inflammation. However, ischemic insult leads to a pathogenic level of IgM binding and complement activation, resulting in inflammation and injury. We investigate the role of self-reactive IgM in the unique setting of transplantation where the donor organ undergoes both cold and warm ischemia and global ischemic insult.

Methods and results: By transplanting hearts from wild-type donor mice into antibody-deficient mice reconstituted with specific self-reactive IgM monoclonal antibodies, we identified neoepitopes expressed after transplantation and demonstrated a key role for IgM recognition of these epitopes in graft injury. With this information, we developed and characterized a therapeutic strategy that exploited the postischemia recognition system of natural antibodies. On the basis of neoepitope identification, we constructed an anti-annexin IV single-chain antibody (scFv) and an scFv linked to Crry, an inhibitor of C3 activation (scFv-Crry). In an allograft transplantation model in which recipients contain a full natural antibody repertoire, both constructs blocked graft IgM binding and complement activation and significantly reduced graft inflammation and injury. Furthermore, scFv-Crry specifically targeted to the transplanted heart and, unlike complement deficiency, did not affect immunity to infection, an important consideration for immunosuppressed transplant recipients.

Conclusions: We identified pathophysiologically important epitopes expressed within the heart after transplantation and described a novel translatable strategy for targeted complement inhibition that has several advantages over currently available approaches.

Keywords: antibodies; complement system proteins; inflammation; ischemia; transplantation.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Annexin A4 / immunology
  • Antibodies, Bispecific / genetics
  • Antibodies, Bispecific / immunology
  • Antibodies, Bispecific / therapeutic use*
  • Antibodies, Monoclonal / genetics
  • Antibodies, Monoclonal / immunology
  • Antibodies, Monoclonal / therapeutic use*
  • Complement Activation
  • Epitopes / immunology
  • Genes, Synthetic
  • Heart Transplantation / adverse effects*
  • Homeodomain Proteins / genetics
  • Immunoglobulin M / deficiency
  • Immunoglobulin M / genetics
  • Immunoglobulin M / immunology
  • Immunoglobulin M / therapeutic use*
  • Male
  • Mice
  • Mice, Inbred BALB C
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Myocardial Reperfusion / adverse effects*
  • Myocardial Reperfusion Injury / immunology
  • Myocardial Reperfusion Injury / prevention & control*
  • Myocardium / immunology
  • Organ Specificity
  • Phospholipids / immunology
  • Receptors, Complement / genetics
  • Receptors, Complement / therapeutic use*
  • Receptors, Complement 3b
  • Recombinant Fusion Proteins / genetics
  • Recombinant Fusion Proteins / immunology
  • Recombinant Fusion Proteins / therapeutic use
  • Self Tolerance / immunology*
  • Single-Chain Antibodies / genetics
  • Single-Chain Antibodies / immunology
  • Single-Chain Antibodies / therapeutic use*
  • Transplantation Tolerance

Substances

  • Annexin A4
  • Antibodies, Bispecific
  • Antibodies, Monoclonal
  • Cr1l protein, mouse
  • Epitopes
  • Homeodomain Proteins
  • Immunoglobulin M
  • Phospholipids
  • Receptors, Complement
  • Receptors, Complement 3b
  • Recombinant Fusion Proteins
  • Single-Chain Antibodies
  • RAG-1 protein