Macrophages and Kidney Transplantation

https://doi.org/10.1016/j.semnephrol.2010.03.009Get rights and content

Summary

Macrophages are present within the transplanted kidney in varying numbers throughout its lifespan. Because of their prominence during acute rejection episodes, macrophages traditionally have been viewed as contributors to T-cell–directed graft injury. With growing appreciation of macrophage biology, it has become evident that different types of macrophages exist within the kidney, subserving a range of functions that include promotion or attenuation of inflammation, participation in innate and adaptive immune responses, and mediation of tissue injury and fibrosis, as well as tissue repair. A deeper understanding of how macrophages accumulate within the kidney and of what factors control their differentiation and function may identify novel therapeutic targets in transplantation.

Section snippets

Phenotypic and Functional Diversity of Monocytes and Macrophages

Monocyte and macrophage biology is reviewed on page 216 in this issue of Seminars in Nephrology1 and will be discussed here only briefly. Circulating bone marrow–derived monocytes exist in subsets that are characterized by unique sets of cell surface markers. Human monocytes may be separated into CD16+ or CD16 subsets with CD16 monocytes expressing the chemokine receptors CCR1 and CCR2. Circulating murine monocytes also may be separated into 2 discrete populations with different chemokine

Macrophage Participation in Kidney IRI and the Importance of IRI in the Development of Acute Rejection

As an essential initial event in the process of kidney transplantation, kidney retrieval, preservation, transportation, and implantation impart an unavoidable episode of IRI. This response has been relatively well characterized in experimental models and is known to be mediated by engagement of endogenous ligands, expressed in response to IRI, by innate immune receptors such as TLRs expressed by kidney parenchymal cells and cells of bone-marrow origin, such as macrophages.16, 17, 18 Evidence

Macrophages in Acute Kidney Allograft Rejection

The initial descriptions of experimental allograft rejection by Brent et al39 in 1958 noted the accumulation of macrophages and T cells in a delayed-type hypersensitivity–like response. In kidney transplantation, a small number of resident tissue macrophages of donor origin are transplanted with the graft.40 Macrophages accumulate within the kidney in response to IRI and then taper after 7 to 14 days to low numbers.41 Although such passenger macrophages may proliferate after implantation,40 the

Role of Macrophages as Mediators of Kidney Inflammation and Damage During Acute Rejection

Several potential roles exist for macrophages during acute rejection including phagocytosis, processing and presentation of antigen to primed CD4+ T cells, proinflammatory cytokine production, effector cell functions causing tissue damage, and finally immune modulation and the promotion of tissue repair. It is likely that such diverse functions are conducted by phenotypically distinct subpopulations of macrophages within the graft.

Anti-Inflammatory and Immunosuppressant Activity of Macrophages Within the Kidney

Macrophages may be deactivated by innate stimuli such as uptake of apoptotic cells, cytokines such as IL-10, TGF-β, and IFN-α/β, as well as therapeutic agents such as glucocorticoids.1, 5, 7, 9, 86 Furthermore, macrophage deactivation may promote differentiation into immunoregulatory macrophages, characterized by low-level expression of MHC class II but high level production of key anti-inflammatory cytokines including IL-10 and TGF-β.1, 7, 9

Although it is highly likely that macrophages are

Macrophages in Chronic Allograft Nephropathy

Although acute rejection is a common event and a significant contributor to graft loss during the first postoperative year, CAN characterized by interstitial fibrosis and tubular atrophy is a far greater cause of graft loss overall.87 The pathogenesis of CAN remains unclear, however, inflammation, mediated via allo-specific (true chronic rejection) and nonspecific (drug toxicity, ischemia) pathways, is a constant feature.88 Macrophage infiltration is a feature of alloimmunity, but may be

Macrophages in Transplant-Associated Lymphangiogenesis

The lymphatic vessels draining the normal kidney are disrupted during organ retrieval with no attempt made to reconnect them surgically to the lymphatic system of the recipient. Early work in dogs suggested that the transplanted kidney effectively becomes reconnected to the systemic lymphatic system within 2 weeks102 and this is important for tubulointerstitial tissue homeostasis.103 More recent work using specific markers for the lymphatic endothelium has shown significant interstitial

Conclusions and Future Prospects

Macrophages are a major presence within the transplanted kidney. Different types of macrophage involvement have been shown at various stages of the allograft response. These include contributions to the innate immune response to renal IRI, effector cell function causing cell damage, and inflammation and the facilitation of adaptive immune responses. In addition, macrophages may promote healing and repair as the graft recovers from acute insults and be involved in the detrimental development of

References (114)

  • R. Girlanda et al.

    Monocyte infiltration and kidney allograft dysfunction during acute rejection

    Am J Transplant

    (2008)
  • M.D. Jose et al.

    Blockade of macrophage colony-stimulating factor reduces macrophage proliferation and accumulation in renal allograft rejection

    Am J Transplant

    (2003)
  • G.A. Bishop et al.

    Immunopathology of renal allograft rejection analyzed with monoclonal antibodies to mononuclear cell markers

    Kidney Int

    (1986)
  • A.B. Magil et al.

    Monocytes and peritubular capillary C4d deposition in acute renal allograft rejection

    Kidney Int

    (2003)
  • T. Fahim et al.

    The cellular lesion of humoral rejection: predominant recruitment of monocytes to peritubular and glomerular capillaries

    Am J Transplant

    (2007)
  • L. Martinez-Pomares et al.

    Antigen presentation the macrophage way

    Cell

    (2007)
  • H. Okamura et al.

    Interleukin-18: a novel cytokine that augments both innate and acquired immunity

    Adv Immunol

    (1998)
  • K. Wyburn et al.

    Interleukin-18 affects local cytokine expression but does not impact on the development of kidney allograft rejection

    Am J Transplant

    (2006)
  • C.A. Dinarello

    Interleukin-1 and interleukin-1 antagonism

    Blood

    (1991)
  • V. Holan et al.

    Nitric oxide as a regulatory and effector molecule in the immune system

    Mol Immunol

    (2002)
  • I. Herrero-Fresneda et al.

    Do alloreactivity and prolonged cold ischemia cause different elementary lesions in chronic allograft nephropathy?

    Am J Pathol

    (2003)
  • L.G. Fine et al.

    Chronic hypoxia as a mechanism of progression of chronic kidney diseases: from hypothesis to novel therapeutics

    Kidney Int

    (2008)
  • F. Geissmann et al.

    Blood monocytes: distinct subsets, how they relate to dendritic cells, and their possible roles in the regulation of T-cell responses

    Immunol Cell Biol

    (2008)
  • C. Auffray et al.

    Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior

    Science

    (2007)
  • J. Savill et al.

    A blast from the past: clearance of apoptotic cells regulates immune responses

    Nat Rev Immunol

    (2002)
  • K.J. Mylonas et al.

    Alternatively activated macrophages elicited by helminth infection can be reprogrammed to enable microbial killing

    J Immunol

    (2009)
  • D.M. Mosser et al.

    Exploring the full spectrum of macrophage activation

    Nat Rev Immunol

    (2008)
  • R. van Furth

    Origin and turnover of monocytes and macrophages

    Curr Top Pathol

    (1989)
  • S. Gordon

    Alternative activation of macrophages

    Nat Rev Immunol

    (2003)
  • J. Banchereau et al.

    Immunobiology of dendritic cells

    Annu Rev Immunol

    (2000)
  • D.M. Underhill et al.

    Dynamic interactions of macrophages with T cells during antigen presentation

    J Exp Med

    (1999)
  • P. Chomarat et al.

    TNF skews monocyte differentiation from macrophages to dendritic cells

    J Immunol

    (2003)
  • G.J. Randolph et al.

    The CD16(+) (FcgammaRIII(+)) subset of human monocytes preferentially becomes migratory dendritic cells in a model tissue setting

    J Exp Med

    (2002)
  • W.P. Pulskens et al.

    Toll-like receptor-4 coordinates the innate immune response of the kidney to renal ischemia/reperfusion injury

    PLoS One

    (2008)
  • J.C. Leemans et al.

    Renal-associated TLR2 mediates ischemia/reperfusion injury in the kidney

    J Clin Invest

    (2005)
  • H. Wu et al.

    TLR4 activation mediates kidney ischemia/reperfusion injury

    J Clin Invest

    (2007)
  • B. Kruger et al.

    Donor Toll-like receptor 4 contributes to ischemia and reperfusion injury following human kidney transplantation

    Proc Natl Acad Sci U S A

    (2009)
  • D.T. Fearon et al.

    The instructive role of innate immunity in the acquired immune response

    Science

    (1996)
  • G. Chalasani et al.

    The allograft defines the type of rejection (acute versus chronic) in the face of an established effector immune response

    J Immunol

    (2004)
  • S.G. Yarlagadda et al.

    Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysis

    Nephrol Dial Transplant

    (2009)
  • D.K. Ysebaert et al.

    Identification and kinetics of leukocytes after severe ischaemia/reperfusion renal injury

    Nephrol Dial Transplant

    (2000)
  • S.K. Jo et al.

    Macrophages contribute to the initiation of ischaemic acute renal failure in rats

    Nephrol Dial Transplant

    (2006)
  • J. Menke et al.

    CSF-1 signals directly to renal tubular epithelial cells to mediate repair in mice

    J Clin Invest

    (2009)
  • S.L. Lin et al.

    Macrophage Wnt7b is critical for kidney repair and regeneration

    Proc Natl Acad Sci U S A

    (2010)
  • C.E. McCoy et al.

    The role of toll-like receptors in macrophages

    Front Biosci

    (2008)
  • H. Wu et al.

    IL-18 contributes to renal damage after ischemia-reperfusion

    J Am Soc Nephrol

    (2008)
  • K.K. Donnahoo et al.

    Early kidney TNF-alpha expression mediates neutrophil infiltration and injury after renal ischemia-reperfusion

    Am J Physiol

    (1999)
  • V.Y. Melnikov et al.

    Impaired IL-18 processing protects caspase-1-deficient mice from ischemic acute renal failure

    J Clin Invest

    (2001)
  • M.L. Kielar et al.

    Maladaptive role of IL-6 in ischemic acute renal failure

    J Am Soc Nephrol

    (2005)
  • D.B. Ascon et al.

    Phenotypic and functional characterization of kidney-infiltrating lymphocytes in renal ischemia reperfusion injury

    J Immunol

    (2006)
  • Cited by (30)

    • Elevated Monocyte Count and Loss of Renal Function in Renal Transplant Patients

      2020, Transplantation Proceedings
      Citation Excerpt :

      Dendritic cells function in cellular rejection, stimulate and activate T cells, and are derived from macrophages [4–6]. One review reported that macrophages play a role in matrix formation and fibrosis development in various disorders ranging from ischemic reperfusion injury to acute rejection [7]. In light of this knowledge, we investigated the effect of peripheral blood monocyte count on post-transplant period kidney function by comparing 2 groups who did and did not receive ATG.

    • ISN Forefronts Symposium 2015: The Diverse Function of Macrophages in Renal Disease

      2016, Kidney International Reports
      Citation Excerpt :

      Recent work has suggested an important role for retinoic acid in modulating macrophage phenotype via the direct inhibition of M1 macrophages and the promotion of tubular cell induction of M2 macrophages, indicating that there are multiple pathways to manipulate macrophage phenotype.60 Macrophages are present in human renal diseases, including diabetes,61,62 polycystic kidney disease, kidney allograft rejection,63 chronic allograft nephropathy,64,65 and acute kidney injury.22,65 Studies have demonstrated a strong association between the extent of macrophage infiltration and functional outcome.66

    • Macrophages in renal transplantation: Roles and therapeutic implications

      2014, Cellular Immunology
      Citation Excerpt :

      Macrophages have long been recognized within the graft during IRI, acute and chronic rejection [5]. Traditionally, these cells were viewed as contributors to T-cell mediated processes such as acute rejection, recruited into the graft under the influence of T cell derived chemokines to promote inflammation, cause tissue injury and act as antigen presenting cells (APCs) [6]. The more recent discovery of Toll-like receptors (TLRs) and their essential role as innate activators of macrophages during organ IRI has led to a growing appreciation of the role of macrophages and innate immunity in allograft responses and highlighted the importance of innate-adaptive cross-talk in the development of adaptive immune responses.

    • Immunology of graft rejection

      2013, Kidney Transplantation-Principles and Practice, Seventh Edition
    • Determination of optimal incubation time for the production of acute phase cytokines ex vivo by peripheral blood mononuclear cells from renal transplant recipients

      2011, Journal of Immunological Methods
      Citation Excerpt :

      However, a biopsy has several drawbacks over a simple blood draw, as mentioned (Danovitch, 2005). There is accumulating evidence that, in addition to T-cell mediated adaptive immunity processes, inflammation originating from the innate immunity plays an active role during allograft rejection and thus could serve as a potential biomarker of graft status (Chadban et al., 2010). We and others have previously reported that 36 to 48 h incubation of PBMCs results in measurable levels of acute phase cytokines in humans (transplantation paper and (De Serres et al., 2011; Bradshaw et al., 2009).

    View all citing articles on Scopus

    Professor Chadban and Dr Wu receive research funding from the National Health and Medical Research Council of Australia; and Dr Hughes is supported by funding from the UK Medical Research Council, Kidney Research UK, and the Genzyme Renal Innovation Program.

    View full text