Elsevier

Clinical Immunology

Volume 130, Issue 1, January 2009, Pages 41-50
Clinical Immunology

REVIEW
The innate immune response in ischemic acute kidney injury

https://doi.org/10.1016/j.clim.2008.08.016Get rights and content

Abstract

Kidney ischemia reperfusion injury is a major cause of morbidity in both allograft and native kidneys. Ischemia reperfusion-induced acute kidney injury is characterized by early, alloantigen-independent inflammation. Major components of the innate immune system are activated and participate in the pathogenesis of acute kidney injury, plus prime the allograft kidney for rejection. Soluble members of innate immunity implicated in acute kidney injury include the complement system, cytokines, and chemokines. Toll-like receptors (TLRs) are also important contributors. Effector cells that participate in acute kidney injury include the classic innate immune cells, neutrophils and macrophages. Recent data has unexpectedly identified lymphocytes as participants of early acute kidney injury responses. In this review, we will focus on immune mediators that participate in the pathogenesis of ischemic acute kidney injury.

Introduction

Acute kidney injury is associated with prolonged hospitalization, higher morbidity and mortality [1]. Ischemia reperfusion injury is a leading cause of acute kidney injury in both transplanted and native kidneys. Ischemia reperfusion-induced acute kidney injury is associated with tubulointerstitial inflammation and a robust inflammatory response to hypoxia and the process of reperfusion [2], [3]. In this review, we will discuss the role of the innate immune system in ischemic acute kidney injury, as well as present recent data on a modulatory role for lymphocytes.

In ischemic acute kidney injury, hypoxic and anoxic cell injuries occur early during the ischemic phase, followed by inflammatory responses in the reperfusion phase (Fig. 1). During reperfusion, blood containing innate immune component flows through ischemic tissues and accentuates injury. It is well established that a robust inflammatory reaction occurs following ischemia reperfusion [3]. Renal ischemia reperfusion induces renal synthesis or activation of pro-inflammatory cytokines and chemokines, and recruits leukocytes into the post-ischemic kidneys—to be reviewed in more detail in following sections. Several reports demonstrating the reno-protective effects of therapy targeting innate immune components such as complements, chemokines (discussed in the following section), or adhesion molecules [4], [5], [6], directly support the role of innate immunity in the pathogenesis of acute kidney injury. Functional impairment of kidney during acute kidney injury, leading to retention of fluids and nitrogenous waste products, further aggravates and sustains inflammation.

The initiation signals activating the innate immune system as well as triggering inflammatory response can be classified into 4 categories: 1) factors passively released from injured cells, 2) factors actively synthesized and secreted from the cells that have undergone ischemia, 3) recognition of altered or injured cell structures, and 4) decreased expression of anti-inflammatory factors by injured cells [7]. In acute kidney injury, both endothelium and tubular epithelium participate in innate immune responses. The signaling responses in tubular epithelium during renal injury, such as signaling through toll-like receptors (TLRs), is quite similar with that during ascending urinary infection [8].

Section snippets

Soluble molecules and membrane-associated receptors

The innate immune response includes soluble molecules such as complement and cytokines. In order to initiate and generate a full-blown innate immune response, TLRs appear required. We will review their roles in ischemic acute kidney injury in this section.

Cellular mediators of innate immunity

Macrophages, neutrophils, natural killer (NK) cells, and dendritic cells are important cells involved in innate immune responses (Table 1). In addition, newer data also implicates lymphocytes during these early injury responses.

Other leukocytes participating in early injury responses (“innate-like”)

There are several minor lymphocytes subsets which express receptors with very limited diversity and do not undergo clonal expansion before responding effectively to the antigens. Therefore, they are known as innate-like lymphocytes (ILLs). Three main classes of ILLs are NK T cells, intraepithelial γδ T cells, and B-1 subset of B cells (B-1 cells) (Table 2).

Conclusion

Robust early inflammatory responses occur in post-ischemic kidneys, facilitating the full expression of tissue damage and organ dysfunction in acute kidney injury. Numerous experimental studies have revealed the importance of innate immune responses following ischemia reperfusion injury. Based on this information about the role of immune component of ischemic acute kidney injury, there is an opportunity to further dissect the underlying mechanisms. It will be important to study how early immune

Acknowledgments

The authors thank Dr. Maria-Teresa Gandolfo for helpful suggestions with manuscript. HR is supported by the US National Institutes of Health and US National Kidney Foundation.

References (96)

  • F. Togel et al.

    Renal SDF-1 signals mobilization and homing of CXCR4-positive cells to the kidney after ischemic injury

    Kidney Int.

    (2005)
  • S.T. Tarzami et al.

    Chemokine expression in myocardial ischemia: MIP-2 dependent MCP-1 expression protects cardiomyocytes from cell death

    J. Mol. Cell Cardiol.

    (2002)
  • X. Che et al.

    Monocyte chemoattractant protein-1 expressed in neurons and astrocytes during focal ischemia in mice

    Brain Res.

    (2001)
  • K. Furuichi et al.

    Chemokine receptor CX3CR1 regulates renal interstitial fibrosis after ischemia–reperfusion injury

    Am. J. Pathol.

    (2006)
  • S.G. Soriano et al.

    Mice deficient in fractalkine are less susceptible to cerebral ischemia–reperfusion injury

    J. Neuroimmunol.

    (2002)
  • K.E. De Greef et al.

    Anti-B7-1 blocks mononuclear cell adherence in vasa recta after ischemia

    Kidney Int.

    (2001)
  • J.W. Celie et al.

    Subendothelial heparan sulfate proteoglycans become major L-selectin and monocyte chemoattractant protein-1 ligands upon renal ischemia/reperfusion

    Am. J. Pathol.

    (2007)
  • V.P. Persy et al.

    Reduced postischemic macrophage infiltration and interstitial fibrosis in osteopontin knockout mice

    Kidney Int.

    (2003)
  • F. Gueler et al.

    Statins attenuate ischemia–reperfusion injury by inducing heme oxygenase-1 in infiltrating macrophages

    Am. J. Pathol.

    (2007)
  • T. Nemoto et al.

    Small molecule selectin ligand inhibition improves outcome in ischemic acute renal failure

    Kidney Int.

    (2001)
  • J.J. Friedewald et al.

    Inflammatory cells in ischemic acute renal failure

    Kidney Int.

    (2004)
  • H. Chiao et al.

    Alpha-melanocyte-stimulating hormone inhibits renal injury in the absence of neutrophils

    Kidney Int.

    (1998)
  • T. Hayama et al.

    Beneficial effect of neutrophil elastase inhibitor on renal warm ischemia–reperfusion injury in the rat

    Transplant. Proc.

    (2006)
  • A.J. Turunen et al.

    Activated protein C reduces graft neutrophil activation in clinical renal transplantation

    Am. J. Transplant.

    (2005)
  • S. Mizuno et al.

    Prevention of neutrophil extravasation by hepatocyte growth factor leads to attenuations of tubular apoptosis and renal dysfunction in mouse ischemic kidneys

    Am. J. Pathol.

    (2005)
  • J.G. Penfield et al.

    Transplant surgery injury recruits recipient MHC class II-positive leukocytes into the kidney

    Kidney Int.

    (1999)
  • C.L. Schlichting et al.

    Renal ischemia–reperfusion injury: new implications of dendritic cell-endothelial cell interactions

    Transplant. Proc.

    (2006)
  • X. Dong et al.

    Resident dendritic cells are the predominant TNF-secreting cell in early renal ischemia–reperfusion injury

    Kidney Int.

    (2007)
  • A. Loverre et al.

    Ischemia–reperfusion injury-induced abnormal dendritic cell traffic in the transplanted kidney with delayed graft function

    Kidney Int.

    (2007)
  • V. Savransky et al.

    Role of the T-cell receptor in kidney ischemia–reperfusion injury

    Kidney Int.

    (2006)
  • E.A. Jones et al.

    The effect of mycophenolate mofetil and polyphenolic bioflavonoids on renal ischemia reperfusion injury and repair

    J. Urol.

    (2000)
  • A. Chandraker et al.

    CD28-b7 blockade in organ dysfunction secondary to cold ischemia/reperfusion injury

    Kidney Int.

    (1997)
  • L.W. Lai et al.

    A sphingosine-1-phosphate type 1 receptor agonist inhibits the early T-cell transient following renal ischemia–reperfusion injury

    Kidney Int.

    (2007)
  • S. Wang et al.

    Decreased renal ischemia–reperfusion injury by IL-16 inactivation

    Kidney Int.

    (2008)
  • R. Thadhani et al.

    Acute renal failure

    N. Engl. J. Med.

    (1996)
  • K. Solez et al.

    The morphology of “acute tubular necrosis” in man: analysis of 57 renal biopsies and a comparison with the glycerol model

    Medicine (Baltimore)

    (1979)
  • K.J. Kelly et al.

    Intercellular adhesion molecule-1-deficient mice are protected against ischemic renal injury

    J. Clin. Invest.

    (1996)
  • M. Takada et al.

    The cytokine-adhesion molecule cascade in ischemia/reperfusion injury of the rat kidney. Inhibition by a soluble P-selectin ligand

    J. Clin. Invest.

    (1997)
  • K. Singbartl et al.

    Protection from ischemia–reperfusion induced severe acute renal failure by blocking E-selectin

    Crit. Care Med.

    (2000)
  • J.M. Thurman et al.

    Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice

    J. Immunol.

    (2003)
  • J.M. Thurman et al.

    Altered renal tubular expression of the complement inhibitor Crry permits complement activation after ischemia/reperfusion

    J. Clin. Invest.

    (2006)
  • J.M. Thurman et al.

    C3a is required for the production of CXC chemokines by tubular epithelial cells after renal ischemia/reperfusion

    J. Immunol.

    (2007)
  • J.M. Thurman et al.

    Treatment with an inhibitory monoclonal antibody to mouse factor B protects mice from induction of apoptosis and renal ischemia/reperfusion injury

    J. Am. Soc. Nephrol.

    (2006)
  • X. Zheng et al.

    Protection of renal ischemia injury using combination gene silencing of complement 3 and caspase 3 genes

    Transplantation

    (2006)
  • M. Moller-Kristensen et al.

    Mannan-binding lectin recognizes structures on ischaemic reperfused mouse kidneys and is implicated in tissue injury

    Scand. J. Immunol.

    (2005)
  • W. Zhou et al.

    Predominant role for C5b-9 in renal ischemia/reperfusion injury

    J. Clin. Invest.

    (2000)
  • B. de Vries et al.

    Complement factor C5a mediates renal ischemia–reperfusion injury independent from neutrophils

    J. Immunol.

    (2003)
  • S. David et al.

    Alternative pathway complement activation induces proinflammatory activity in human proximal tubular epithelial cells

    Nephrol. Dial. Transplant.

    (1997)
  • Cited by (289)

    View all citing articles on Scopus
    View full text