Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jan 18.
Published in final edited form as: Kidney Int Suppl. 2010 Dec;(119):S18–S21. doi: 10.1038/ki.2010.417

Innate immunity and cardiac allograft rejection

Timothy M Millington 1, Joren C Madsen 1
PMCID: PMC3261228  NIHMSID: NIHMS345544  PMID: 21116311

Abstract

The development of immunosuppressive drugs to control adaptive immune responses has led to the success of heart transplantation as a therapy for end-stage heart failure. However, these agents are largely ineffective in suppressing components of the innate immune system. This distinction has gained clinical significance as mounting evidence now indicates that innate immune responses have important roles in the acute and chronic rejection of cardiac allografts including cardiac allograft vasculopathy (CAV). Whereas clinical interest in natural killer (NK) cells was once largely confined to the field of bone marrow transplantation, recent findings suggest that these cells can also participate in the acute rejection of cardiac allografts and in the development of CAV. Stimulation of Toll-like receptors (TLRs), another important component of innate immunity, by endogenous ligands released in response to ischemia/reperfusion is now known to cause an inflammatory milieu favorable to graft rejection. Finally, new data indicate that activation of complement is linked to acute rejection and CAV. In summary, the conventional wisdom that the innate immune system is of little importance in whole-organ transplantation is no longer tenable. The addition of strategies that target TLRs, NK cells, and complement will be necessary to prevent CAV completely and to eventually achieve long-term tolerance to cardiac allografts.

Keywords: complement, heart transplant, innate immunity, natural killer cells, Toll-like receptors


Although more than 85,000 heart transplants have been performed worldwide since 1982 and the transplant half-life has improved to 10 years,1 the procedure remains a palliation rather than a cure. The need for lifelong immunosuppression with its attendant side effects of infection and malignancy, coupled with the high incidence of cardiac allograft vasculopathy (CAV)—43% by the end of the eighth post-transplant year2—contributes to a steady linear increase in mortality by 3.5% each year, continuing for more than 15 years after transplantation.

Increasing the understanding of the adaptive immune system has permitted the development of drugs to prevent or treat acute cellular rejection, leading to significant improvement in survival in the first year after transplantation.2 However, critical events immediately after heart transplantation, including graft infiltration by natural killer (NK) cells, activation of Toll-like receptors (TLRs), and complement deposition may set the stage for the development of graft injury and chronic rejection, ultimately leading to graft loss. Understanding the innate immune response to the cardiac allograft is therefore critical to maximizing the therapeutic benefits of cardiac transplantation.

NK Cells

The principal cellular component of the innate immune system is the NK cell, a lymphoid cell that is capable of responding to target cells without previous sensitization. NK cells can be activated by the absence (‘missing’) of self major histocompatibility complex molecules on their cellular targets and by the stimulation of receptors capable of identifying a limited number of viral and tumor antigens. When the combination of stimulation and inhibition received by the NK cell leads to activation, the outcome is target cell lysis by perforin and initiation of inflammation by cytokine release, principally interferon-γ (IFN-γ). Until recently, NK cells were believed not to acquire adaptive memory or contribute to the rejection of solid allografts. However, this established paradigm has been challenged by a number of reports that suggest a more complex role for this lymphocyte subset in whole-organ transplantation.

It is clear that NK cells are active in the early stages of allograft rejection. In the first few days after allogeneic heart transplantation, the majority of infiltrating lymphocytes are NK cells.3 In murine recipients of cardiac allografts, expression of the NK cell-activating receptor NKG2D and its ligands, including retinoic acid early inducible (RAE-1) and minor histocompatibility antigen H60, is upregulated from 3 to 5 days after transplantation.4 Expression increases with time as rejection develops, and only a modest effect is seen in the recipients of syngeneic grafts, suggesting that this receptor–ligand interaction could have a role in stimulating rejection.

Early evidence that NK cells could have a role in the acute rejection of cardiac allografts originated with studies using co-stimulation-deficient (CD28−/−) mice. Although blockade of the CD28-B7 co-stimulatory interaction with anti-CD154 leads to tolerance of cardiac allografts in mice, CD28-deficient mice remain able to reject cardiac allografts through a CD8-mediated process.5 CD 28−/− mice can, however, be made to accept cardiac allografts by depletion of NK1.1+ T cells.6 In CD28−/− mice, a subpopulation of NK cells homes to allogeneic (but not syngeneic) grafts after transplantation. Antibody-mediated blockade of the activating receptor NKG2D prolongs the survival of cardiac allografts in CD28−/− mice from 21.3 to 70.1 days.7 This finding suggested that NK cells could facilitate antigen-specific CD8+ T-lymphocyte proliferation leading to graft rejection, either through the direct action of secreted cytokines or by promoting dendritic cell maturation.8

Although studies in CD28−/− mice suggest that NK cells participate in acute rejection by promoting the effects of alloreactive T lymphocytes rather than by some intrinsic capability of NK cells to directly reject solid organ allografts (reviewed in Kitchens et al.9), under the right conditions, mice lacking T and B lymphocytes (Rag−/−) are able to reject allogeneic skin through an NK-mediated process.10 When skin allografts were placed on Rag−/− mice, NK cells infiltrated the graft but did not lead to rejection. However, when the animals were treated with interleukin (IL)-15, a powerful stimulator of NK cells, the allografts were rejected as the NK population expanded. In Rag−/− γc −/− animals, which also lack NK cells, no rejection occurred. Finally, when IL-15 was withdrawn, the ability of NK cells to reject the graft was lost. These studies counter conventional wisdom and suggest that IL-15-activated NK cells have the capability to reject major histocompatibility complex disparate allografts directly, without contributing to the adaptive immune system.

NK cells have also been implicated in the development of CAV in experiments using a parental-to-F1 strain combination. Transplanting mouse heart from parental donors to F1 hybrid recipients avoids activation of the adaptive immune system and isolates responses mediated by the innate immune system.11 Solid organs transplanted in this manner were accepted indefinitely without immunosuppression as expected, as there is no host T- or B-cell response against the donor. However, when parental allografts were removed 56 days after transplantation, 19 of 22 had developed advanced CAV without evidence of acute rejection. IFN-γ-deficient recipients of parental-to-F1 hybrid transplants did not develop CAV. These data strongly suggest that NK cells can contribute to the formation of vascular lesions of CAV through an IFN-γ-dependent pathway.

In a global sense, previous studies suggest that the time-honored distinctions between the adaptive immune system and innate immune system are beginning to blur. This is best exemplified by recent studies showing that NK cells can exhibit immunological memory, the sine qua non of the adaptive immune system. Sun et al.12 expanded mouse NK cells carrying a receptor specific for cytomegalovirus (Ly49H) by infecting them with cytomegalovirus. They demonstrated that a portion of these expanded Ly49H-positive cells can mount a secondary expansion after a repeated stimulus. ‘Memory’ NK cells retain this property after adoptive transfer to a naive animal. There is also evidence for antigen-independent development of an NK memory-like subset. NK cells can be expanded in vitro with IL-12 and IL-18, with IL-15 as a survival factor and adoptively transferred into Rag−/− mice, which lack T and B cells.13 These expanded NK cells resemble host NK cells, but result in higher levels of IFN-γ than do naive NK cells in response to a second stimulus 1–3 weeks later, although they do not have enhanced cytotoxicity. The discovery of memory-like NK suggests that CAV-free acceptance of cardiac grafts may require therapies to block the development of NK memory or deplete existing populations of previously sensitive NK memory cells.

There is only limited evidence for a central role for NK cells in acute rejection of cardiac allografts in human patients. One recent report 14 compared NK cell numbers in peripheral blood and endomyocardium in 20 patients with acute cellular rejection (grade 3a) with that of 19 stable patients (grade 0). A marked reduction in NK cells was detected in the blood of rejecting patients with an associated increase in CD16+ NK cells in graft biopsies, raising the possibility that during rejection NK cells travel from the blood to the graft.

TLRs

TLRs are among the best-studied components of the innate immune system. This ancient group of transmembrane proteins is expressed on epithelial cells, dendritic cells, macrophages, and T and B lymphocytes. TLRs recognize conserved ligands such as lipopolysaccharide on Gram-negative bacteria and similar structures on other pathogenic microbes. There is evidence that TLRs are also activated by endogenous ligands,15 some of which are released following ischemia/reperfusion (I/R) injury.

There is now evidence that the I/R injury, which inevitably accompanies transplantation of a cold-preserved cardiac allograft, is associated with the development of accelerated atherosclerosis.16 This process may be mediated by TLRs. For instance, tissue damage releases endogenous proteins such as the extracellular matrix polysaccharide hyaluronan,17 which activates dendritic cells by binding to TLR4. Messenger molecules, including high-mobility group box 1 and heat-shock cognate protein 70, are also released from the heart after I/R injury, which in turn initiate a TLR4-dependent inflammatory response.18 In uninjured mouse hearts, heat-shock cognate protein 70 is confined to the cytoplasm but is released into the coronary vessels after a period of warm ischemia and reperfusion. Proinflammatory molecules tumor necrosis factor (TNF)-α, IL-1, and IL-6 are increased by I/R and by recombinant heat-shock cognate protein 70. This effect is diminished by antibody blockade of heat-shock cognate protein 70 and is absent in TLR4-deficient mice. A similar phenomenon is observed with high-mobility group box 1, which is released within 30 min of ligation of the left anterior descending artery. Inhibition of high-mobility group box 1 limits I/R injury, whereas treatment with recombinant high-mobility group box 1 leads to an enhanced inflammatory response.19 Elevations of TLR4 mRNA, TNF-α, and IL-6 are also seen after I/R injury in rats.20

The inflammatory response triggered by I/R-mediated TLR4 impairs myocardial function. Wild-type mice display significant increases in TNF-α and IL-1 after I/R injury, which may be responsible for the associated depression of left ventricular diastolic pressure and dP/Dtmax.21 This theory is supported by the fact that deficiencies in TLR4, TNF-α, and IL-1 are cardioprotective and that treatment with exogenous TNF-α and IL-1 restores the harmful effects of I/R in TLR4-deficient mice. Syngeneic heart transplants in TLR4-deficient mice are associated with reduced neutrophil infiltration and lower graft and serum levels of inflammatory cytokines including TNF-α, IL-6, monocyte chemoattractant protein-1, and IL-1.22

The effects of TLR activation on the cardiac allograft may be long lasting and may contribute to the development of CAV. In a series of human heart transplant recipients, expression of TLR4 (and its targets IL-12 and TNF-α) was significantly elevated in patients with allograft endothelial dysfunction, which is a predictor of subsequent development of CAV.23

TLR activation inhibits the development of tolerance to cardiac allografts. B6 mice spontaneously develop tolerance to cardiac grafts from major histocompatibility complex class II-disparate bm12 donors without immunosuppression, but rejection is restored by treatment with the TLR agonist CpG. CpG induces Th1 differentiation of naive T cells and attenuates suppression by Tregs.24 Tolerance can be induced experimentally by blockade of CD28-B7 co-stimulation with anti-CD154 antibody, but treatment with CpG permits rejection to occur and inhibits the accumulation of Tregs in the cardiac allograft.25

Finally, there is a link between NK-cell effects and TLRs. The activation of NK cells through the NKG2D receptor–ligand interaction may be initiated by TLRs. Various TLRs are found on T and B lymphocytes, dendritic cells, macrophages, and epithelial cells, and bind to both exogenous and endogenous ligands.15 On exposure to lipopolysaccharide, monocytes and macrophages produce NKG2D ligands including RAE-126 and major histocompatibility complex class I-related chain A.27 Monocytes thus activated can, in the presence of IL-2, stimulate NK cells to secrete IFN-γ. When dendritic cells are depleted, NK cell activation by the TLR ligand CpG is inhibited.28

Complement

When acute cellular rejection is present, there is increased expression of complement genes by graft-infiltrating leukocytes. Analysis of samples from human heart transplant recipients demonstrates higher transcript numbers of complement factor B, C3, and properdin, and C3a receptor and C5a receptor, as well as other genes potentially associated with rejection, including CD3, IFN-γ, perforin, and granzyme B.29 Deficiency of decay-accelerating factor, a naturally occurring inhibitor of complement activation, leads to antibody-independent acceleration of graft rejection, possibly through increased proliferation of alloreactive CD8 + T cells.30

I/R injury causes host complement proteins to bind the graft endothelium, possibly initiating a process of inflammation, coagulation, and permanent tissue damage. A series of cardiac graft biopsy specimens in the first several weeks after transplantation demonstrated that deposition of C4d and C3d was associated with histological evidence of peritransplant ischemic injury; patients with complement deposition were more likely to demonstrate repeated episodes of rejection on later biopsies.31 C4d deposition has recently been linked to the development of CAV in the first year after transplantation.32

Defects in complement activation may protect against cellular rejection. A ‘minibody’ antagonist of C5 given before cardiac transplantation in rats leads to lower levels of TNF-α and reduced cardiomyocyte death.33 In humans, a naturally occurring defect in mannose-binding lectin, a protein involved in complement activation, is associated with a decrease in the number of episodes of cardiac allograft rejection.34

Together, these findings demonstrate a newfound importance for complement as a mediator of both the acute and chronic rejection of cardiac allografts.

Conclusion

Although conventional immunosuppressive drugs, including antiproliferative agents and calcineurin inhibitors, effectively suppress the adaptive immune system, they have little, if any, effect on components of the innate immune system. Recognition that the innate immune system has a major role in acceptance or rejection of a cardiac allograft demands that strategies aimed at blocking the activation of NK cells, the response to TLRs, and the deposition of complement be incorporated into protocols aimed at preventing CAV and inducing long-term tolerance to cardiac allografts.

Acknowledgments

This work was supported in part by the National Heart, Lung, and Blood Institute (PO1HL018646, RO1HL093131) and the National Institute of Allergy and Infectious Disease (U19AI066705) of the National Institutes of Health and the Roche Organ Transplantation Research Foundation (ROTRF). TMM is the recipient of an International Society of Heart & Lung Transplantation (ISHLT) research fellowship.

Footnotes

Disclosure: The authors declared no competing interests.

References

  • 1.Taylor D, Stehlik J, Edwards L, et al. Registry of the International Society for Heart and Lung Transplantation: Twenty-sixth Official Adult Heart Transplant Report—2009. J Heart Lung Transplant. 2009;28:1007–1022. doi: 10.1016/j.healun.2009.08.014. [DOI] [PubMed] [Google Scholar]
  • 2.Schmauss D, Weis M. Cardiac allograft vasculopathy: recent developments. Circulation. 2008;117:2131–2141. doi: 10.1161/CIRCULATIONAHA.107.711911. [DOI] [PubMed] [Google Scholar]
  • 3.Dresske B, Zhu X, Herwartz C, et al. The time pattern of organ infiltration and distribution of natural killer cells and macrophages in the course of acute graft rejection after allogeneic heart transplantation in the rat. Transplant Proc. 1997;29:1715. doi: 10.1016/s0041-1345(97)00026-2. [DOI] [PubMed] [Google Scholar]
  • 4.Feng L, Ke N, Ye Z, et al. Expression of NKG2D and its ligand in mouse heart allografts may have a role in acute rejection. Transplant Proc. 2009;41:4332–4339. doi: 10.1016/j.transproceed.2009.08.060. [DOI] [PubMed] [Google Scholar]
  • 5.Szot G, Zhou P, Rulifson I, et al. Different mechanisms of cardiac allograft rejection in wildtype and CD28-deficient mice. Am J Transplant. 2001;1:38–46. doi: 10.1034/j.1600-6143.2001.010108.x. [DOI] [PubMed] [Google Scholar]
  • 6.Maier S, Tertilt C, Chambron N, et al. Inhibition of natural killer cells results in acceptance of cardiac allografts in CD28−/− mice. Nat Med. 2001;7:557–562. doi: 10.1038/87880. [DOI] [PubMed] [Google Scholar]
  • 7.Kim J, Chang C, Hayden T, et al. The activating immunoreceptor NKG2D and its ligands are involved in allograft transplant rejection. J Immunol. 2007;179:6416–6420. doi: 10.4049/jimmunol.179.10.6416. [DOI] [PubMed] [Google Scholar]
  • 8.McNerney M, Lee K, Zhou P, et al. Role of natural killer cell subsets in cardiac allograft rejection. Am J Transplant. 2006;6:505–513. doi: 10.1111/j.1600-6143.2005.01226.x. [DOI] [PubMed] [Google Scholar]
  • 9.Kitchens W, Uehara S, Chase C, et al. The changing role of natural killer cells in solid organ rejection and tolerance. Transplantation. 2006;81:811–817. doi: 10.1097/01.tp.0000202844.33794.0e. [DOI] [PubMed] [Google Scholar]
  • 10.Kroemer A, Xiao X, Degauque N, et al. The innate NK cells, allograft rejection, and a key role for IL-15. J Immunol. 2008;180:7818–7826. doi: 10.4049/jimmunol.180.12.7818. [DOI] [PubMed] [Google Scholar]
  • 11.Uehara S, Chase C, Kitchens W, et al. NK cells can trigger allograft vasculopathy: the role of hybrid resistance in solid organ allografts. J Immunol. 2005;175:3424–3430. doi: 10.4049/jimmunol.175.5.3424. [DOI] [PubMed] [Google Scholar]
  • 12.Sun JC, Beilke JN, Lanier LL. Adaptive immune features of natural killer cells. Nature. 2009;457:557–561. doi: 10.1038/nature07665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cooper M, Elliott J, Keyel P, et al. Cytokine-induced memory-like natural killer cells. Proc Natl Acad Sci USA. 2009;106:1915–1919. doi: 10.1073/pnas.0813192106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Fildes J, Bishop P, Mitchell J, et al. 422: Natural killer cells deplete from peripheral blood and increase in myocardial tissue during acute rejection following heart transplantation. J Heart Lung Transplant. 2008;27:S212–S213. doi: 10.1016/j.healun.2007.11.571. [DOI] [PubMed] [Google Scholar]
  • 15.Chao W. Toll-like receptor signaling: a critical modulator of cell survival and ischemic injury in the heart. Am J Physiol Heart Circ Physiol. 2009;296:H1–H12. doi: 10.1152/ajpheart.00995.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gaudin P, Rayburn B, Hutchins G, et al. Peritransplant injury to the myocardium associated with the development of accelerated arteriosclerosis in heart transplant recipients. Am J Surg Pathol. 1994;18:338–346. doi: 10.1097/00000478-199404000-00002. [DOI] [PubMed] [Google Scholar]
  • 17.Shirali A, Goldstein D. Activation of the innate immune system by the endogenous ligand hyaluronan. Curr Opin Organ Transplant. 2008;13:20–25. doi: 10.1097/MOT.0b013e3282f3df04. [DOI] [PubMed] [Google Scholar]
  • 18.Zou N, Ao L, Cleveland J, et al. Critical role of extracellular heat shock cognate protein 70 in the myocardial inflammatory response and cardiac dysfunction after global ischemia-reperfusion. Am J Physiol Heart Circ Physiol. 2008;294:H2805–H2813. doi: 10.1152/ajpheart.00299.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Andrassy M, Volz H, Igwe JC, et al. High-mobility group box-1 in ischemia-reperfusion injury of the heart. Circulation. 2008;117:3216–3226. doi: 10.1161/CIRCULATIONAHA.108.769331. [DOI] [PubMed] [Google Scholar]
  • 20.Yang J, Yang J, Ding J, et al. Sequential expression of TLR4 and its effects on the myocardium of rats with myocardial ischemia-reperfusion injury. Inflammation. 2008;31:304–312. doi: 10.1007/s10753-008-9079-x. [DOI] [PubMed] [Google Scholar]
  • 21.Cha J, Wang Z, Ao L, et al. Cytokines link toll-like receptor 4 signaling to cardiac dysfunction after global myocardial ischemia. Ann Thorac Surg. 2008;85:1678–1685. doi: 10.1016/j.athoracsur.2008.01.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kaczorowski D, Nakao A, Mollen K, et al. Toll-like receptor 4 mediates the early inflammatory response after cold ischemia/reperfusion. Transplantation. 2007;84:1279–1287. doi: 10.1097/01.tp.0000287597.87571.17. [DOI] [PubMed] [Google Scholar]
  • 23.Methe H, Zimmer E, Grimm C, et al. Evidence for a role of toll-like receptor 4 in development of chronic allograft rejection after cardiac transplantation. Transplantation. 2004;78:1324–1331. doi: 10.1097/01.tp.0000137930.40597.03. [DOI] [PubMed] [Google Scholar]
  • 24.Porrett P, Yuan X, LaRosa D, et al. Mechanisms underlying blockade of allograft acceptance by TLR ligands. J Immunol. 2008;181:1692–1699. doi: 10.4049/jimmunol.181.3.1692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chen L, Wang T, Zhou P, et al. TLR engagement prevents transplantation tolerance. Am J Transplant. 2006;6:2282–2291. doi: 10.1111/j.1600-6143.2006.01489.x. [DOI] [PubMed] [Google Scholar]
  • 26.Hamerman J, Ogasawara K, Lanier L. Cutting edge: toll-like receptor signaling in macrophages induces ligands for the NKG2D receptor. J Immunol. 2004;172:2001–2005. doi: 10.4049/jimmunol.172.4.2001. [DOI] [PubMed] [Google Scholar]
  • 27.Kloss M, Decker P, Baltz K, et al. Interaction of monocytes with NK cells upon toll-like receptor-induced expression of the NKG2D ligand MICA. J Immunol. 2008;181:6711–6719. doi: 10.4049/jimmunol.181.10.6711. [DOI] [PubMed] [Google Scholar]
  • 28.Hochweller K, Striegler J, Hämmerling G, et al. A novel CD11c.DTR transgenic mouse for depletion of dendritic cells reveals their requirement for homeostatic proliferation of natural killer cells. Eur J Immunol. 2008;38:2776–2783. doi: 10.1002/eji.200838659. [DOI] [PubMed] [Google Scholar]
  • 29.Keslar K, Rodriguez E, Tan C, et al. Complement gene expression in human cardiac allograft biopsies as a correlate of histologic grade of injury. Transplantation. 2008;86:1319–1321. doi: 10.1097/TP.0b013e3181889831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pavlov V, Raedler H, Yuan S, et al. Donor deficiency of decay-accelerating factor accelerates murine T cell-mediated cardiac allograft rejection. J Immunol. 2008;181:4580–4589. doi: 10.4049/jimmunol.181.7.4580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Baldwin W, Samaniego-Picota M, Kasper E, et al. Complement deposition in early cardiac transplant biopsies is associated with ischemic injury and subsequent rejection episodes. Transplantation. 1999;68:894–900. doi: 10.1097/00007890-199909270-00024. [DOI] [PubMed] [Google Scholar]
  • 32.Poelzl G, Ullrich R, Huber A, et al. Capillary deposition of the complement fragment C4d in cardiac allograft biopsies is associated with allograft vasculopathy. Transpl Int. 2005;18:313–317. doi: 10.1111/j.1432-2277.2004.00037.x. [DOI] [PubMed] [Google Scholar]
  • 33.Ferraresso M, Macor P, Valente M, et al. Posttransplant ischemia-reperfusion injury in transplanted heart is prevented by a minibody to the fifth component of complement. Transplantation. 2008;86:1445–1451. doi: 10.1097/TP.0b013e31818a68e2. [DOI] [PubMed] [Google Scholar]
  • 34.Fildes J, Shaw S, Walker A, et al. Mannose-binding lectin deficiency offers protection from acute graft rejection after heart transplantation. J Heart Lung Transplant. 2008;27:1353–1356. doi: 10.1016/j.healun.2008.08.011. [DOI] [PubMed] [Google Scholar]

RESOURCES