Macrophages in Renal Disease

BRIEF REVIEW www.jasn.org Macrophages in Renal Disease Yiping Wang and David C.H. Harris Centre for Transplant and Renal Research, Westmead Millenni...
Author: Eustacia Tyler
2 downloads 0 Views 2MB Size
BRIEF REVIEW

www.jasn.org

Macrophages in Renal Disease Yiping Wang and David C.H. Harris Centre for Transplant and Renal Research, Westmead Millennium Institute, The University of Sydney, Sydney, New South Wales, Australia

ABSTRACT Macrophages have heterogeneous phenotypes as they exercise their twofold role in the development and recovery of renal diseases. Some subpopulations of macrophages (M1) have a pathogenic function in renal inflammation, making them a logical target for elimination. Alternatively, M2 macrophage subpopulations resolve inflammation and repair injury, making them a potential therapeutic tool against renal injury. Here, we summarize recent findings regarding macrophage plasticity, and the various strategies for targeting or utilizing macrophages to treat renal disease. We highlight, in particular, the potential of renoprotective M2 macrophages to resolve inflammation and repair the kidney. J Am Soc Nephrol 22: 21–27, 2011. doi: 10.1681/ASN.2010030269

Macrophage infiltration is a common feature of most human chronic kidney disease. A critical question is whether these macrophages cause or exacerbate renal injury. Correlations between the degree of macrophage infiltration and severity of renal injury in humans, and in temporal studies in rodents, suggest an effector function for macrophages.1 This correlation in humans with chronic kidney disease is more of a generality2 but is well studied in specific diseases including IgA nephropathy and lupus nephritis.3 However, such strong correlations do not assure the link is causal. Proof of causality comes from animal studies that demonstrate induction of injury by pathogenic mediators of macrophages, improvement of renal injury and function by depletion of macrophages, and acceleration of renal injury by repletion of macrophages. Studies over many years demonstrate that macrophages express many cytotoxic moieties, including proteolytic enzymes, reactive oxygen and nitrogen species, and proinflammatory cytokines and chemokines.4 For example, macrophages J Am Soc Nephrol 22: 21–27, 2011

isolated from nephritic glomeruli of rats and rabbits generated reactive oxygen species and nitric oxide.5–7 Glomerular macrophages produce TNF␣ and IL-1␤ in animal models of both lupus nephritis and nephrotoxic serum nephritis.8 –12 Immunohistologic studies demonstrate that macrophages infiltrating the kidneys of patients and rodents with focal necrotizing glomerulonephritis express MHC class II molecules, inducible nitric oxide synthase, and inflammatory cytokines.5,13 Activated macrophages can also induce epithelial-mesenchymal transition (EMT) forming fibroblasts by secreting matrix metalloproteinase 9.14 A variety of strategies to deplete macrophages also prove that they contribute to renal injury.15 Macrophage depletion using irradiation, antimacrophage sera, lipid-encapsulated drugs, or blockade of macrophage chemokine receptors reduces proteinuria and structural injury in models including antiglomerular basement membrane (GBM) disease, puromycin aminonucleoside nephrosis, and Heymann nephritis.16 –19 Another powerful approach to clarify

the role of macrophages in renal disease has been the use of adoptive transfer techniques. For example, transferred macrophages induce proteinuria and mesangial cell proliferation in anti-GBM nephritis.20 Transfer of macrophages activated by lipopolysaccharide, but not resting macrophages, also exacerbate renal injury in adriamycin nephrosis.21 In the latter studies, transferred macrophages accumulate progressively in sites of injury and as few as 10,000 macrophages per mouse are sufficient to exacerbate disease. These data suggest specific targeting of activated macrophages is likely to be a more effective therapeutic approach than inhibition of all macrophages subpopulations.

MACROPHAGES AS A TARGET IN RENAL DISEASE

Numerous studies over the past decade provide compelling evidence that macrophages cause tissue injury. Since the classic depletion studies of macrophages in renal disease by Holdsworth et al. in the 1980s,17 a variety of strategies including disruption of macrophage recruitment into kidney and genetic modification of macrophage activity by gene Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. Yiping Wang, Centre for Transplant and Renal Research, Westmead Millennium Institute, The University of Sydney, Sydney, NSW, Australia. Phone: 61-2-98456877; Fax: 61-296339351; E-mail: [email protected] Copyright © 2011 by the American Society of Nephrology

ISSN : 1046-6673/2201-21

21

BRIEF REVIEW

www.jasn.org

intervention have been employed to reduce macrophage infiltration. Macrophage Depletion

Pathogenic macrophages can be depleted directly with antimacrophage serum in experimental glomerulonephritis,17 and partially reduced in adriamycin nephropathy using ED7, an antibody directed against CD11b/CD18 integrin, which is expressed by macrophages.22 Blockade of colony-stimulating factor receptor on macrophages, c-fms, also reduces macrophage accumulation in unilateral ureteral obstruction23 and murine diabetic nephropathy24 by markedly reducing macrophage proliferation. Treatment with liposomal clodronate also reduces the accumulation of F4/80⫹ macrophages and renal fibrosis in obstruction.25 Targeting monocyte CD11b with diphtheria toxin receptor (DTR) in a transgenic mouse (CD11b-DTR) also effectively halts progression of crescentic glomerulonephritis.26 Disruption of Macrophage Recruitment

Inhibition of CX3CR1, the receptor for the chemokine fractalkine (CX3CL1), markedly reduces macrophage infiltration and injury in a model of crescentic glomerulonephritis.27 Anti-CC chemokine ligand (CCL) 2 antibody reduces macrophage infiltration, crescent formation, and proteinuria in rats with nephrotoxic serum nephritis19,28,29 or anti-thy 1.1 nephritis.30 In adriamycin nephropathy, macrophage recruitment and functional and structural renal injury are reduced by combined CCL2 and CCL5 DNA vaccination31 or CCL2 DNA vaccine modified to increase its immunogenicity.32 However, strategies targeting chemokines or adhesion molecules have not been uniformly effective. Treatment with anti-CCL2 antibody in Wistar Kyoto (WKY) rats with anti-GBM disease did not reduce crescent formation and proteinuria, despite early reduction of macrophage infiltration.33 Blockade of CCL5 reduces renal injury in anti-thy 1.1 nephritis in rats and in murine nephrotoxic nephritis,34,35 but exacerbates in22

flammation in murine immune-complex nephritis, despite a reduction in the number of infiltrating macrophages.36 Moreover, antibody blockade of intercellular adhesion molecule-1 (ICAM-1) counter-receptor reduces macrophage infiltration in WKY rats with nephrotoxic nephritis;37 however, administration of anti-CD18 antibody against counter-receptor to ICAM-1 and -2 does not reduce macrophage infiltration in Lewis rats with nephrotoxic nephritis (Figure 1 and Table 1A).38 Genetic Alteration of Macrophage Activity

Inhibition or enhancement of macrophage gene expression can be achieved by gene silencing or transfer using antisense oligonucleotides (ODN) to silence specific macrophage genes. In rats with nephrotoxic serum nephritis, intravenous ODN targeting NF-␬B reduces macrophage infiltration, proteinuria, and proinflammatory cytokine expression.39 Blockade of ICAM-1 with ODN also reduces macrophage infiltration and attenuates reperfusion injury in the rat.40 However, inhibition of CCL2 using antiCCL2 spiegelmers reduces macrophage infiltration, but does not improve renal pathology in mice with Alport nephropathy.41 Specific genes have also been transferred to alter macrophage activation or inhibit signaling pathways. Genetically engineered macrophages can de-

liver genes specifically to sites of inflammation by following natural chemotactic signals. For example, macrophages transduced with adenovirus expressing IL-1ra reduce renal injury in both obstructive nephropathy and nephrotoxic nephritis.42,43 Rees and co-workers showed that macrophages transduced with IL-4 or IL-10 were able to reduce histologic injury and albuminuria in rats with nephrotoxic nephritis,44,45 but IL-10 expressing macrophages were more effective than those expressing IL-4.46 The same group transferred inhibitor of protein ␬B (I␬B) into macrophages to block NF-␬B, thereby reducing renal injury in rats with nephrotoxic nephritis (Figure 2 and Table 1B).47 In summary, depletion of macrophages or disruption of macrophage recruitment reduces renal macrophage accumulation and injury in diseased kidney. Absence of specific surface markers for macrophages has meant that macrophage depletion strategies lack specificity. The conflicting results of blockade of macrophage chemokines or adhesion molecules suggest the pivotal importance of specific macrophage infiltration pathways and states of activation as well as the exact nature of the renal injury. Whereas the specific inhibition of macrophage genes through antisense or decoy ODN has proven successful, their utility is limited by inhibitory effects, which are shortlived (minutes to days), and by off-target

Block chemokine and adhesion molecules CCL-2, -3, -5, CX3CL1, MIF, VLA-4, ICAM-1, VCAM

and their receptors

Target macrophage surface markers CD11b, CD11b/CD18, F4/80

Silence macrophage genes ODN, sRNA, etc.

Monocytes

Block macrophage signaling pathways NF-κb, JNK, MAPK

Inhibit macrophage proliferation Anti-c-fms, cyclophosphamide

Induce macrophage apoptosis Liposomal clodronate

Figure 1. Strategies for targeting macrophages to treat kidney disease. Renal injury may be reduced by strategies aimed at reducing the number and activity of effector macrophages.

Journal of the American Society of Nephrology

J Am Soc Nephrol 22: 21–27, 2011

www.jasn.org

BRIEF REVIEW

Table 1. Published studies of macrophage strategies for treating renal disease Method A. Macrophage As Target antimacrophage sera ED7 antibody anti–c-fms liposomal clodronate CD11b-DTR anti-CX3CR1 anti-CCL2 CCL2 and CCL5 DNA vaccine modified CCL2 DNA vaccine anti-CCL5 anti–ICAM-1, anti-CD18 NF-␬B decoy ODN ICAM-1 ODN CCL2 spiegelmers B. Macrophage As Tool alternatively activated macrophages (M2a) alternatively activated macrophages (M2c) IL-1ra transfected macrophages IL-4 transfected macrophages IL-10 transfected macrophages macrophages transduced with I␬B

Animal Model

Reference(s)

Anti-GBM disease (rabbit) AN (rat) UUO (rat); diabetic nephropathy (db/db mouse) Anti-GBM disease Crescentic glomerulonephritis (mouse) Crescentic glomerulonephritis (WKY rat) Anti-GBM disease (rat and WKY rat); crescentic nephritis (mouse); anti-thy 1.1 nephritis (mouse) AN (rat) AN (rat) Anti-thy 1.1 nephritis (mouse); nephrotoxic nephritis (mouse); immune-complex nephritis (mouse) Nephrotoxic nephritis (WKY rat) Crescentic glomerulonephritis (rat) Renal ischaemia-reperfusion injury (rat) Alport nephropathy (mouse)

17 22 23,24 25 26 27 19,28–30,33

31 32 34–36

37,38 39 40 41

AN (mouse) STZ-induced DN (BALB/c and eNOS⫺/⫺ mouse) AN (mouse) Anti-GBM disease (rat); UUO (rat) Nephrotoxic nephritis (rat) Nephrotoxic nephritis (rat) Nephrotoxic nephritis (rat)

57–59 60 42,43 44 45,46 47

AN, adriamycin nephropathy; DN, diabetic nephropathy; UUO, unilateral ureteral obstruction; WKY, Wistar-Kyoto.

reactions with other genes with sequence similarity. Macrophages can be genetically engineered to deliver anti-inflammatory genes or inhibit signaling pathways, providing long-lasting expression and sitespecific migration after chemotactic signals. However, substantial hurdles to the future clinical application of genetic transfer using macrophages include unregulated gene expression and concerns about the unproven safety of viral vectors.

DIVERSE ROLES OF MACROPHAGES

Macrophages comprise a heterogeneous population of cells, with diverse functions and phenotypic plasticity. Although commonly recognized for their pathogenic role in renal inflammation and fibrosis, macrophages also play critical roles in wound healing, in tissue remodeling and repair, and in immune regulation. Macrophages that secrete anti-inflammatory cytokines and J Am Soc Nephrol 22: 21–27, 2011

promote wound healing and tissue remodeling have been referred to as alternatively activated macrophages (M2). By their secretion of trophic factors and anti-inflammatory cytokines, M2 macrophages resolve inflammation and reduce

4 ILMonocyte

, IL-1

3

IL1

0, T G



M2 programmed ex vivo

M2a

Immune complex + LPS M2c

Virus Iκb

injury.48,49 Macrophages are also able to fuse with themselves and other cell types, thereby providing the potential to regenerate specialized cells of the kidney.50 Three subsets of alternatively activated macrophages have been proposed:

M2b

Mechanisms –Reduce infiltration of host macrophages –Deactivate lost effector macrophages –Inhibit CD4 cell proliferation –Reduce CD8 cell cytoxicity –Induce Tregs (M2c)

–Increase renal IL-4 and IL-10 –Decrease renal TNFα, IL-1 and IFNγ –Reduce infiltration and activation of macrophages

IL-10 IL-4

IL-1ra

Genetically modified macrophages

Figure 2. Mechanisms underlying beneficial effects of macrophages modified by exposure to cytokines ex vivo, or by transfection with anti-inflammatory genes. The mechanisms explaining the protective effect of macrophages against renal injury involve inhibition of effector macrophages, effector lymphocytes and proinflammatory cytokines, and the promotion of regulatory T lymphocytes and anti-inflammatory cytokines. Macrophages in Renal Disease

23

BRIEF REVIEW

www.jasn.org

those exposed to IL-4 and IL-13, called M2a or wound-healing macrophages; those exposed to immune complexes and IL-1␤ or LPS, termed M2b; and those exposed to IL-10 and TGF␤ or glucocorticoids, termed M2c or regulatory macrophages. Although classically activated, pathogenic macrophages (M1) have been characterized well, the differences and inter-relationships among subsets of M2 in terms of specific markers and functions have not been delineated clearly. In mice with unilateral ureteric obstruction, macrophage depletion during progressive inflammatory fibrosis ameliorate scarring and reduce myofibroblast numbers, whereas macrophage depletion during the recovery phase leads to a failure of matrix degradation and persistent scarring, revealing the presence of two distinct macrophage phenotypes, M1 and M2.53 Another study showed that FTY720, an immunosuppressive modulator of sphingosine 1-phosphate receptor, decreases M2 macrophages in 5 of 6 nephrectomized rats.54 We have been unable to demonstrate a unique subset of macrophages that exhibit features of M2 macrophages in normal kidney, in contrast to the demonstration of M2 macrophages in normal intestine55 and lung.56 Whether M2 macrophages exist in diseased kidneys awaits demonstration in both humans and in animal models. Just as the presence of M1 macrophages in damaged kidney does not prove their pathogenic role, so the demonstration of M2 macrophages does not prove that they are mitigating damage. 51,52

MACROPHAGES AS A TOOL IN RENAL DISEASES

Although extensive in vitro studies demonstrate that M2 macrophages secrete anti-inflammatory cytokines such as IL-10 and TGF␤ and express inhibitory molecules such as indoleamine 2,3-dioxygenase, mannose receptor, and arginase, whether M2 macrophages have an anti-inflammatory role in in vivo renal disease is open to question. If M2 macrophages are to be used as a therapeutic 24

tool in vivo, two conditions must be met: a stable phenotype and the ability to access inflamed sites or local organ draining lymph nodes. Our data demonstrate that after cytokine modulation suppressive features of M2 macrophages are maintained for up to 4 weeks in vitro.57 M2 macrophages express greater numbers of chemokine receptors than resting macrophages and readily accumulate in inflamed kidney and renal draining lymph nodes. Given their phenotypic stability and ability to access diseased kidney, M2 macrophages have been tested for their ability to modify renal disease in animal models. Our group first examined the role of M2 macrophages in SCID mice with adriamycin nephropathy, a model of human focal segmental glomerulosclerosis, to determine their direct effects on injury in the absence of endogenous T and B cells. In that study, macrophages were isolated from spleen and incubated with IL-4 and IL-13 ex vivo to induce an M2 phenotype (M2a). These macrophages were then infused into SCID mice with adriamycin nephropathy, where they reduced histologic and functional injury.57 The protective effect of M2a macrophages was associated with reduced accumulation and downregulated expression of inflammatory cytokines of endogenous effector macrophages. The regulatory effects of M2 macrophages were target-specific and sustained. Subsequently, we showed a similar protective effect of M2a macrophages against renal structural and functional injury in immunocompetent mice with adriamycin nephropathy. Our group has also investigated the protective effects of M2a macrophages in another model, that of murine streptozotocin-induced (STZinduced) diabetes. Transfused M2a macrophages accumulate progressively in kidneys for at least 10 weeks after STZ. Kidneys from diabetic mice transfused with M2a macrophages have less glomerular hypertrophy, tubular atrophy, interstitial expansion, and interstitial fibrosis than did diabetic mice not transfused.58 Similarly, M2a macrophage transfusion of diabetic endothelial nitric oxide synthase null (eNOS⫺/⫺) mice resulted in

Journal of the American Society of Nephrology

less arteriolar hyalinosis and glomerulosclerosis, and a lower systolic BP than in diabetic eNOS⫺/⫺ mice who were not transfused (Figure 2 and Table 1B).59

THE POTENCY OF DIFFERENT SUBSETS AND DIFFERENT SOURCES OF M2 MACROPHAGES

Another subset of M2 macrophages produced by exposure to cytokines IL-10 and TGF␤ (M2c) is known to exhibit anti-inflammatory cytokine production and suppressive functions in vitro; however, its therapeutic efficacy is unproven and relative potency in comparison to M2a macrophages unknown. Studies demonstrating the efficacy of M2c macrophages in reducing injury in murine adriamycin nephropathy have been published recently.60 The relative efficacy of M2a and M2c macrophages were compared in adriamycin nephropathy (unpublished). M2c macrophages display a greater protective effect than M2a macrophages against tubular atrophy, interstitial expansion, and proteinuria. This greater potency of M2c than M2a macrophages could relate to the high-level expression by M2c but not M2a macrophages of the regulatory co-stimulation molecule, B7-H4, and the ability to induce T regulatory cells from CD4⫹CD25⫺ T lymphocytes.61 In the above studies, M2 macrophages, produced ex vivo from resting splenic macrophages, ameliorate adriamycin nephropathy. However, the importance of the organ of origin of M2 macrophages is unknown. Therefore, the effects of splenic M2 and bone marrow M2 macrophages induced ex vivo by IL4/IL-13 (M2a) were compared in adriamycin nephropathy. In vitro, M2a from spleen or bone marrow both showed high-level expression of IL-10 and TGF␤ and regulatory molecules, mannose receptor, and arginase. Likewise, they suppress effector macrophages and cytotoxic CD8⫹ T cells with similar efficacy. However, unlike M2a macrophages derived from spleen, those from bone marrow do not protect against renal structural or functional injury.62 To investigate the failure of J Am Soc Nephrol 22: 21–27, 2011

www.jasn.org

bone marrow M2a macrophages to protect against nephropathy, transfused macrophages (labeled with DiI) were separated from inflamed kidney and examined by cell sorting. Expression of anti-inflammatory cytokines and regulatory molecules by transfused bone marrow M2a macrophages, but not by transfused splenic M2a macrophages, is dramatically reduced in inflamed kidney. The loss of suppressive function of bone marrow M2a macrophages is linked to their proliferation within inflamed kidney. M2a macrophages from bone marrow, but not from spleen, proliferate strongly in kidney, and dividing cells do not express the regulatory phenotype of M2 macrophages.

PROTECTIVE MECHANISMS OF M2 MACROPHAGES

The mechanisms underlying the renoprotective effects of M2 macrophages involve effects on activated macrophages, CD4 T cells, and CD8 T cells. M2 macrophages are also able to suppress classically activated M1 macrophages, which exhibit reduced production of proinflammatory cytokines and iNOS in the presence of M2 macrophages.60 Macrophage-mediated protection could also involve phagocytosis of damaged cells and cell debris. Our demonstration that M2 macrophages protect against injury in immunodeficient mice with adriamycin nephropathy suggests they act independently of lymphocytes.57 M2 macrophages produce anti-inflammatory cytokines that inhibit function and proliferation of effector lymphocytes.63 Both M2a and M2c macrophages are able to inhibit the proliferation of CD4⫹ T cells in vitro.60 Moreover, this inhibition is partially blocked by neutralizing antibodies against IL-10, TGF␤, or the co-stimulatory molecule B7-H4 and requires cell-to-cell contact. In vitro and in vivo, M2c, but not M2a, macrophages convert CD4⫹CD25⫺ T cells to Foxp3⫹ cells. The mechanism underlying conversion of naive T cells to T regulatory cells by M2c macrophages depends on high-level expression of B7-H4 by M2c macrophages. Both M2a and M2c J Am Soc Nephrol 22: 21–27, 2011

macrophages are able to suppress CD8⫹ T cell–mediated toxicity to tubular cells (Figure 2 and Table 1 B).

POTENTIAL DAMAGING EFFECTS OF M2 MACROPHAGES AND THE IMPLICATIONS FOR THERAPY

Macrophages, unlike T cells, which undergo irreversible differentiation upon stimulation, can retain phenotypic plasticity and respond to different environmental signals. Studies indicate that the phenotype of macrophages change over time; for example, macrophages in the earliest stages of cancer resemble classically activated M1 macrophages, yet with tumor growth, those macrophages develop a regulatory phenotype.64 Similarly, macrophages in adipose tissue of nonobese humans have a wound-healing phenotype, whereas those in obese individuals have a proinflammatory phenotype.65 It is not clear whether phenotypic deviation occurs in transfused anti-inflammatory macrophages used therapeutically. In our previous study, macrophage phenotype after initial ex vivo cytokine modulation was stable in vitro and little changed in vivo.57 Recently, we examined macrophages 1 to 3 weeks after transfusion in an immuncompetent model of adriamycin nephropathy; the phenotype of transfused macrophages did drift, but not toward distinct M1 or M2 phenotypes.60 M2 macrophages express high levels of TGF␤, a growth factor linked to renal fibrosis. Macrophage-derived TGF␤ promotes fibrogenesis by paracrine activation of matrix-producing myofibroblasts and tubular epithelial cell transition into myofibroblasts.66 However, recent studies in mice with adriamycin nephropathy treated with M2 macrophages show a reduction instead of promotion of renal fibrosis.57 Interestingly, transfused M2 macrophages gradually decrease their secretion of TGF␤ so that transfused M2 macrophages are persistently antiinflammatory but decreasingly profibrotic with progression of disease.60 Nevertheless, the possible profibrotic

BRIEF REVIEW

effect of M2 macrophages in late stages of chronic kidney disease is unclear. In vivo proliferation of transfused M2 macrophages is another potential obstacle to therapeutic application. M2 macrophages separated from bone marrow, but not spleen, proliferate in vitro for up to 3 weeks and in kidneys of mice with adriamycin nephropathy.60 This proliferation of M2 macrophages associates with loss of their protective phenotype. FUTURE PERSPECTIVES

Before macrophages can become a therapeutic target or tool for human kidney disease, there needs to be a much greater understanding of the biology of these plastic, phenotypically diverse cells. For example, future studies need to examine the specific phenotype of M2 macrophages in various renal diseases, their interaction with dendritic cells and other intrinsic kidney cells, and their potential to fuse with renal cells to generate new tissue. Macrophage modulation ex vivo to produce protective macrophages is a relatively straightforward and effective approach for treating experimental inflammatory renal disease, and such therapeutic macrophages are target-specific, effective, and sustained in their action. However, relative in vivo efficacy of different subsets of M2 macrophages and their dependence on unique features of various kidney diseases remain unclear. Also, the optimal dose, timing, and frequency of M2 macrophages administration need definition. M2 macrophages are effective when given before or soon after the onset of renal injury, whereas their effect in advanced disease is unproven. One of several hurdles to their consideration as a therapeutic tool for humans is their potency. Potential strategies for enhancing their potency include optimizing culture methods with different cytokine combinations and T regulatory cell contact, potentiation of regulatory gene expression, and antigen exposure to induce antigen specificity. DISCLOSURES None.

Macrophages in Renal Disease

25

BRIEF REVIEW

www.jasn.org

REFERENCES 1. Lan HY, Paterson DJ, Atkins RC: Initiation and evolution of interstitial leukocytic infiltration in experimental glomerulonephritis. Kidney Int 40: 425– 433, 1991 2. Ferrario F, Castiglione A, Colasanti G, Barbiano di Belgioioso G, Bertoli S, D’Amico G: The detection of monocytes in human glomerulonephritis. Kidney Int 28: 513–519, 1985 3. Bohle A, Wehrmann M, Bogenschutz O, Batz C, Vogl W, Schmitt H, Muller CA, Muller GA: The long-term prognosis of the primary glomerulonephritides. A morphological and clinical analysis of 1747 cases. Pathol Res Pract 188: 908 –924, 1992 4. Rodriguez-Iturbe B, Pons H, Herrera-Acosta J, Johnson RJ: Role of immunocompetent cells in nonimmune renal diseases. Kidney Int 59: 1626 –1640, 2001 5. Cook HT, Smith J, Salmon JA, Cattell V: Functional characteristics of macrophages in glomerulonephritis in the rat. O2- generation, MHC class II expression, and eicosanoid synthesis. Am J Pathol 134: 431–437, 1989 6. Boyce NW, Tipping PG, Holdsworth SR: Glomerular macrophages produce reactive oxygen species in experimental glomerulonephritis. Kidney Int 35: 778 –782, 1989 7. Cattell V, Largen P, de Heer E, Cook T: Glomeruli synthesize nitrite in active Heymann nephritis; the source is infiltrating macrophages. Kidney Int 40: 847– 851, 1991 8. Tipping PG, Lowe MG, Holdsworth SR: Glomerular interleukin 1 production is dependent on macrophage infiltration in anti-GBM glomerulonephritis. Kidney Int 39: 103–110, 1991 9. Matsumoto K, Dowling J, Atkins RC: Production of interleukin 1 in glomerular cell cultures from patients with rapidly progressive crescentic glomerulonephritis. Am J Nephrol 8: 463– 470, 1988 10. Matsumoto K: Production of interleukin-1 by glomerular macrophages in nephrotoxic serum nephritis. Am J Nephrol 10: 502–506, 1990 11. Tipping PG, Leong TW, Holdsworth SR: Tumor necrosis factor production by glomerular macrophages in anti-glomerular basement membrane glomerulonephritis in rabbits. Lab Invest 65: 272–279, 1991 12. Boswell JM, Yui MA, Burt DW, Kelley VE: Increased tumor necrosis factor and IL-1 beta gene expression in the kidneys of mice with lupus nephritis. J Immunol 141: 3050 – 3054, 1988 13. Noronha IL, Kruger C, Andrassy K, Ritz E, Waldherr R: In situ production of TNF-alpha, IL-1 beta and IL-2R in ANCA-positive glomerulonephritis. Kidney Int 43: 682–692, 1993 14. Tan TK, Zheng G, Hsu TT, Wang Y, Lee VW, Tian X, Wang Y, Cao Q, Wang Y, Harris DC: Macrophage matrix metalloproteinase-9 mediates epithelial-mesenchymal transition in vitro in murine renal tubular cells. Am J Pathol 176: 1256 –1270, 2010

26

15. Nikolic-Paterson DJ, Lan LH, Atkins RC: Macrophages in immune renal injury. In: Immunologic Renal Disease, 2nd Ed., edited by Neilson EG, Couser WG, Philadelphia, Lippincott, Williams & Wilkins, 2001, pp 609 – 632 16. Diamond JR, Pesek-Diamond I: Sublethal Xirradiation during acute puromycin nephrosis prevents late renal injury: Role of macrophages. Am J Physiol 260: F779 –F786, 1991 17. Holdsworth SR, Neale TJ, Wilson CB: Abrogation of macrophage-dependent injury in experimental glomerulonephritis in the rabbit. Use of an antimacrophage serum. J Clin Invest 68: 686 – 698, 1981 18. Hara M, Batsford SR, Mihatsch MJ, BitterSuermann D, Vogt A: Complement and monocytes are essential for provoking glomerular injury in passive Heymann nephritis in rats. Terminal complement components are not the sole mediators of proteinuria. Lab Invest 65: 168 –179, 1991 19. Tang WW, Qi M, Warren JS: Monocyte chemoattractant protein 1 mediates glomerular macrophage infiltration in anti-GBM Ab GN. Kidney Int 50: 665– 671, 1996 20. Ikezumi Y, Hurst LA, Masaki T, Atkins RC, Nikolic-Paterson DJ: Adoptive transfer studies demonstrate that macrophages can induce proteinuria and mesangial cell proliferation. Kidney Int 63: 83–95, 2003 21. Wang Y, Wang Y, Cao Q, Zheng G, Lee VW, Zheng D, Li X, Tan TK, Harris DC: By homing to the kidney, activated macrophages potently exacerbate renal injury. Am J Pathol 172: 1491–1499, 2008 22. Wang Y, Mahajan D, Tay YC, Bao S, Spicer T, Kairaitis L, Rangan GK, Harris DC: Partial depletion of macrophages by ED7 reduces renal injury in Adriamycin nephropathy. Nephrology (Carlton) 10: 470 – 477, 2005 23. Le Meur Y, Tesch GH, Hill PA, Mu W, Foti R, Nikolic-Paterson DJ, Atkins RC: Macrophage accumulation at a site of renal inflammation is dependent on the M-CSF/c-fms pathway. J Leukoc Biol 72: 530 –537, 2002 24. Lim AK, Ma FY, Nikolic-Paterson DJ, Thomas MC, Hurst LA, Tesch GH: Antibody blockade of c-fms suppresses the progression of inflammation and injury in early diabetic nephropathy in obese db/db mice. Diabetologia 52: 1669 –1679, 2009 25. Kitamoto K, Machida Y, Uchida J, Izumi Y, Shiota M, Nakao T, Iwao H, Yukimura T, Nakatani T, Miura K: Effects of liposome clodronate on renal leukocyte populations and renal fibrosis in murine obstructive nephropathy. J Pharmacol Sci 111: 285–292, 2009 26. Duffield JS, Tipping PG, Kipari T, Cailhier JF, Clay S, Lang R, Bonventre JV, Hughes J: Conditional ablation of macrophages halts progression of crescentic glomerulonephritis. Am J Pathol 167: 1207–1219, 2005 27. Feng L, Chen S, Garcia GE, Xia Y, Siani MA, Botti P, Wilson CB, Harrison JK, Bacon KB:

Journal of the American Society of Nephrology

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

Prevention of crescentic glomerulonephritis by immunoneutralization of the fractalkine receptor CX3CR1 rapid communication. Kidney Int 56: 612– 620, 1999 Lloyd CM, Dorf ME, Proudfoot A, Salant DJ, Gutierrez-Ramos JC: Role of MCP-1 and RANTES in inflammation and progression to fibrosis during murine crescentic nephritis. J Leukoc Biol 62: 676 – 680, 1997 Wada T, Yokoyama H, Furuichi K, Kobayashi KI, Harada K, Naruto M, Su SB, Akiyama M, Mukaida N, Matsushima K: Intervention of crescentic glomerulonephritis by antibodies to monocyte chemotactic and activating factor (MCAF/MCP-1). FASEB J 10: 1418 –1425, 1996 Wenzel U, Schneider A, Valente AJ, Abboud HE, Thaiss F, Helmchen UM, Stahl RA: Monocyte chemoattractant protein-1 mediates monocyte/macrophage influx in antithymocyte antibody-induced glomerulonephritis. Kidney Int 51: 770 –776, 1997 Wu H, Wang Y, Tay YC, Zheng G, Zhang C, Alexander SI, Harris DC: DNA vaccination with naked DNA encoding MCP-1 and RANTES protects against renal injury in adriamycin nephropathy. Kidney Int 67: 2178 –2186, 2005 Zheng G, Wang Y, Xiang SH, Tay YC, Wu H, Watson D, Coombes J, Rangan GK, Alexander SI, Harris DC: DNA vaccination with CCL2 DNA modified by the addition of an adjuvant epitope protects against “nonimmune” toxic renal injury. J Am Soc Nephrol 17: 465– 474, 2006 Fujinaka H, Yamamoto T, Takeya M, Feng L, Kawasaki K, Yaoita E, Kondo D, Wilson CB, Uchiyama M, Kihara I: Suppression of antiglomerular basement membrane nephritis by administration of anti-monocyte chemoattractant protein-1 antibody in WKY rats. J Am Soc Nephrol 8: 1174 –1178, 1997 Panzer U, Thaiss F, Zahner G, Barth P, Reszka M, Reinking RR, Wolf G, Helmchen U, Stahl RA: Monocyte chemoattractant protein-1 and osteopontin differentially regulate monocytes recruitment in experimental glomerulonephritis. Kidney Int 59: 1762– 1769, 2001 Lloyd CM, Minto AW, Dorf ME, Proudfoot A, Wells TN, Salant DJ, Gutierrez-Ramos JC: RANTES and monocyte chemoattractant protein-1 (MCP-1) play an important role in the inflammatory phase of crescentic nephritis, but only MCP-1 is involved in crescent formation and interstitial fibrosis. J Exp Med 185: 1371–1380, 1997 Anders HJ, Frink M, Linde Y, Banas B, Wornle M, Cohen CD, Vielhauer V, Nelson PJ, Grone HJ, Schlondorff D: CC chemokine ligand 5/RANTES chemokine antagonists aggravate glomerulonephritis despite reduction of glomerular leukocyte infiltration. J Immunol 170: 5658–5666, 2003 Kawasaki K, Yaoita E, Yamamoto T, Tamatani T, Miyasaka M, Kihara I: Antibodies

J Am Soc Nephrol 22: 21–27, 2011

www.jasn.org

38.

39.

40.

41.

42.

43.

44.

45.

46.

against intercellular adhesion molecule-1 and lymphocyte function-associated antigen-1 prevent glomerular injury in rat experimental crescentic glomerulonephritis. J Immunol 150: 1074 –1083, 1993 Wu X, Tiwari AK, Issekutz TB, Lefkowith JB: Differing roles of CD18 and VLA-4 in leukocyte migration/activation during anti-GBM nephritis. Kidney Int 50: 462– 472, 1996 Tomita N, Morishita R, Lan HY, Yamamoto K, Hashizume M, Notake M, Toyosawa K, Fujitani B, Mu W, Nikolic-Paterson DJ, Atkins RC, Kaneda Y, Higaki J, Ogihara T: In vivo administration of a nuclear transcription factor-kappaB decoy suppresses experimental crescentic glomerulonephritis. J Am Soc Nephrol 11: 1244 –1252, 2000 Kiew LV, Munavvar AS, Law CH, Azizan AN, Nazarina AR, Sidik K, Johns EJ: Effect of antisense oligodeoxynucleotides for ICAM-1 on renal ischaemia-reperfusion injury in the anaesthetised rat. J Physiol 557: 981–989, 2004 Clauss S, Gross O, Kulkarni O, Avila-Ferrufino A, Radomska E, Segerer S, Eulberg D, Klussmann S, Anders HJ: Ccl2/Mcp-1 blockade reduces glomerular and interstitial macrophages but does not ameliorate renal pathology in collagen4A3-deficient mice with autosomal recessive Alport nephropathy. J Pathol 218: 40 – 47, 2009 Yamagishi H, Yokoo T, Imasawa T, Mitarai T, Kawamura T, Utsunomiya Y: Genetically modified bone marrow-derived vehicle cells site specifically deliver an anti-inflammatory cytokine to inflamed interstitium of obstructive nephropathy. J Immunol 166: 609–616, 2001 Yokoo T, Ohashi T, Utsunomiya Y, Kojima H, Imasawa T, Kogure T, Hisada Y, Okabe M, Eto Y, Kawamura T, Hosoya T: Prophylaxis of antibody-induced acute glomerulonephritis with genetically modified bone marrow-derived vehicle cells. Hum Gene Ther 10: 2673–2678, 1999 Wilson HM, Stewart KN, Brown PA, Anegon I, Chettibi S, Rees AJ, Kluth DC: Bone-marrow-derived macrophages genetically modified to produce IL-10 reduce injury in experimental glomerulonephritis. Mol Ther 6: 710 –717, 2002 Kluth DC, Ainslie CV, Pearce WP, Finlay S, Clarke D, Anegon I, Rees AJ: Macrophages transfected with adenovirus to express IL-4 reduce inflammation in experimental glomerulonephritis. J Immunol 166: 4728 – 4736, 2001 Kluth DC, Erwig LP, Rees AJ: Multiple facets

J Am Soc Nephrol 22: 21–27, 2011

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

of macrophages in renal injury. Kidney Int 66: 542–557, 2004 Wilson HM, Chettibi S, Jobin C, Walbaum D, Rees AJ, Kluth DC: Inhibition of macrophage nuclear factor-kappaB leads to a dominant anti-inflammatory phenotype that attenuates glomerular inflammation in vivo. Am J Pathol 167: 27–37, 2005 Herbert DR, Holscher C, Mohrs M, Arendse B, Schwegmann A, Radwanska M, Leeto M, Kirsch R, Hall P, Mossmann H, Claussen B, Forster I, Brombacher F: Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology. Immunity 20: 623– 635, 2004 Pull SL, Doherty JM, Mills JC, Gordon JI, Stappenbeck TS: Activated macrophages are an adaptive element of the colonic epithelial progenitor niche necessary for regenerative responses to injury. Proc Natl Acad Sci U S A 102: 99 –104, 2005 Vignery A: Osteoclasts and giant cells: Macrophage-macrophage fusion mechanism. Int J Exp Pathol 81: 291–304, 2000 Martinez FO, Sica A, Mantovani A, Locati M: Macrophage activation and polarization. Front Biosci 13: 453– 461, 2008 Mosser DM, Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8: 958 –969, 2008 Nishida M, Okumura Y, Fujimoto S, Shiraishi I, Itoi T, Hamaoka K: Adoptive transfer of macrophages ameliorates renal fibrosis in mice. Biochem Biophys Res Commun 332: 11–16, 2005 Schaier M, Vorwalder S, Sommerer C, Dikow R, Hug F, Gross ML, Waldherr R, Zeier M: Role of FTY720 on M1 and M2 macrophages, lymphocytes, and chemokines in 5/6 nephrectomized rats. Am J Physiol Renal Physiol 297: F769 –F780, 2009 Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B: Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol 8: 1086 –1094, 2007 Bedoret D, Wallemacq H, Marichal T, Desmet C, Quesada Calvo F, Henry E, Closset R, Dewals B, Thielen C, Gustin P, de Leval L, Van Rooijen N, Le Moine A, Vanderplasschen A, Cataldo D, Drion PV, Moser M, Lekeux P, Bureau F: Lung interstitial macrophages alter dendritic cell functions to prevent airway allergy in mice. J Clin Invest 119: 3723–3738, 2009

BRIEF REVIEW

57. Wang Y, Wang YP, Zheng G, Lee VW, Ouyang L, Chang DH, Mahajan D, Coombs J, Wang YM, Alexander SI, Harris DC: Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney Int 72: 290 –299, 2007 58. Zheng D, Wang Y, Lee VW, Cao Q, Zheng G, Sun Y, Harris DC: Regulatory macrophages protect against renal injury in murine streptozotocin-induced diabetes. Nephrology (Carlton) 13: A138, 2008 59. Lee VWS, Zheng D, Wang Y, Alexander SI, Harris DCH: Alternatively-activated macrophages, but not regulatory T lymphocytes, ameliorate renal injury in diabetic endothelial nitric oxide synthase knockout mice. Presented in abstract form at the annual meeting of the American Society of Nephrology; October 27 through November 1, 2009; San Diego, CA 60. Cao Q, Wang Y, Zheng D, Sun Y, Wang Y, Lee VW, Alexander SI, Harris DC: IL-10/ TGFb-modified macrophages induce regulatory T cells and protect against murine Adriamycin nephrosis. J Am Soc Nephrol 21: 939 –942, 2010 61. Cao Q, Wang Y, Zheng D, Wang Y, Lee VM, Zheng G, Alexander SI, Harris DC: Discrete functions of protective macrophages determine their relative therapeutic effecacy in chronic kidney disease. Nephrology (Carlton) 14: A16, 2009 62. Cao Q, Zheng D, Sun Y, Wang Y, Lee VW, Zheng G, Alexander SI, Harris DC: Impaired alternative activated macrophages from bone marrow in treatment of CKD: Associations with their proliferation in inflamed kidney. Presented in abstract form at the annual meeting of the American Society of Nephrology; October 27 through November 1, 2009; San Diego, CA 63. Schebesch C, Kodelja V, Muller C, Hakij N, Bisson S, Orfanos CE, Goerdt S: Alternatively activated macrophages actively inhibit proliferation of peripheral blood lymphocytes and CD4⫹ T cells in vitro. Immunology 92: 478 – 486, 1997 64. Sica A, Bronte V: Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Invest 117: 1155–1166, 2007 65. Lumeng CN, Bodzin JL, Saltiel AR: Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175–184, 2007 66. Eddy AA: Progression in chronic kidney disease. Adv Chronic Kidney Dis 12: 353–365, 2005

Macrophages in Renal Disease

27