IRF4 Deficiency Abrogates Lupus Nephritis ... - Semantic Scholar

2 downloads 0 Views 3MB Size Report
Apr 5, 2011 - of systemic lupus and lupus nephritis, but the contribution of IRF4, which has ... less, IRF4 deficiency completely protected these mice from ...
BASIC RESEARCH

www.jasn.org

IRF4 Deficiency Abrogates Lupus Nephritis Despite Enhancing Systemic Cytokine Production Maciej Lech,* Marc Weidenbusch,* Onkar P. Kulkarni,* Mi Ryu,* Murthy Narayana Darisipudi,* Heni Eka Susanti,* Hans-Willi Mittruecker,†‡ Tak W. Mak,‡ and Hans-Joachim Anders*

BASIC RESEARCH

*Department of Nephrology, Medizinische Poliklinik, University of Munich, Munich, Germany; †Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; and ‡The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, Princess Margaret Hospital, Toronto, Ontario, Canada

ABSTRACT The IFN-regulatory factors IRF1, IRF3, IRF5, and IRF7 modulate processes involved in the pathogenesis of systemic lupus and lupus nephritis, but the contribution of IRF4, which has multiple roles in innate and adaptive immunity, is unknown. To determine a putative pathogenic role of IRF4 in lupus, we crossed Irf4-deficient mice with autoimmune C57BL/6-(Fas)lpr mice. IRF4 deficiency associated with increased activation of antigen-presenting cells in C57BL/6-(Fas)lpr mice, resulting in a massive increase in plasma levels of TNF and IL-12p40, suggesting that IRF4 suppresses cytokine release in these mice. Nevertheless, IRF4 deficiency completely protected these mice from glomerulonephritis and lung disease. The mice were hypogammaglobulinemic and lacked antinuclear and anti-dsDNA autoantibodies, revealing the requirement of IRF4 for the maturation of plasma cells. As a consequence, Irf4-deficient C57BL/6(Fas)lpr mice neither developed immune complex disease nor glomerular activation of complement. In addition, lack of IRF4 impaired the maturation of Th17 effector T cells and reduced plasma levels of IL-17 and IL-21, which are cytokines known to contribute to autoimmune tissue injury. In summary, IRF4 deficiency enhances systemic inflammation and the activation of antigen-presenting cells but also prevents the maturation of plasma cells and effector T cells. Because these adaptive immune effectors are essential for the evolution of lupus nephritis, we conclude that IRF4 promotes the development of lupus nephritis despite suppressing antigen-presenting cells. J Am Soc Nephrol 22: 1443–1452, 2011. doi: 10.1681/ASN.2010121260

Systemic autoimmunity in systemic lupus erythematosus (SLE) involves a polyclonal expansion of lymphocytes that are autoreactive to multiple nuclear autoantigens. This process can cause a broad spectrum of clinical manifestations ranging from mild fever, skin rashes, and arthralgia to severe inflammation of kidney, lungs, or brain.1 The pathogenesis of SLE is based on variable combinations of genetic polymorphisms that promote loss of tolerance or tissue inflammation.2,3 For example, certain genes impair lymphocyte apoptosis and the clearance of dying cells via opsonization, phagocytosis, and digestion of self-DNA, which all increase the release of nuclear particles from secondary neJ Am Soc Nephrol 22: 1443–1452, 2011

crotic lymphocytes and expose them to the immune system.4 Another group of susceptibility genes enhances the immune recognition of self nucleic acids by Toll-like receptors (TLRs) in Received December 13, 2010. Accepted April 5, 2011. M.L. and M.W. contributed equally to the manuscript. Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. Hans-Joachim Anders, Medizinische Poliklinik, Universita¨t Mu¨nchen, Pettenkoferstr. 8a, D-80336 Mu¨nchen, Germany. Phone: 49-89-218075855; Fax: 49-8951603379; E-mail: [email protected] Copyright © 2011 by the American Society of Nephrology

ISSN : 1046-6673/2208-1443

1443

BASIC RESEARCH

www.jasn.org

dendritic cells (DCs), which increases the production of of IRF4 in innate and adaptive immunity, we hypothesized a type I IFN5,6 and eventually the expansion of autoreactive functional contribution of IRF4 to SLE and lupus nephritis. lymphocytes.7 A third class of genetic lupus risk factors af- To test this concept, we generated Irf4-deficient C57BL/6lpr/lpr fects tissue inflammation.4 (B6lpr) mice and compared the phenotype with that of wildIFN-regulatory factors (IRFs) form a group of transcription type B6lpr mice, an autoimmune mouse strain that develops factors that have the potential to contribute to all of the afore- lupus autoantibodies and SLE manifestations in kidneys mentioned pathomechanisms of SLE. IRF-1 is a proinflamma- and lungs.27 tory transcription factor that triggers the expression of proinflammatory cytokines in tubular epithelial cells and immune cells in the postischemic kidney8 as well as in mesangial cells during lupus nephritis of MRL-(Fas)lpr mice.9 IRF3 and IRF7 mediate type I IFN production upon immune recognition of viral and endogenous nucleic acids in DCs,10 including TLR7 signaling, which was shown to be an essential pathway in SLE.7,11–13 IRF5 is required for immune cell maturation and for TLR signaling, two mechanisms that contribute to SLE and lupus nephritis of Fc␥RIIB⫺/⫺Yaa or Fc␥RIIB⫺/⫺ mice14 and in lupus secondary to pristane injection.15 Unlike other IRFs, IRF4 is not regulated by IFNs and its expression is restricted to immune cells.16 IRF4 has multiple regulatory functions in adaptive immunity.16,17 For example, IRF4 is required for the maturation of B and T cells,18 plasma cell maturation and Ig isotype switching,19 the ability of regulatory T cells to suppress Th2 responses,20 and the induction of Th17 T cells.21,22 IRF4 also regulates innate immunity because it is required for M2 macrophage polarization.23 IRF4 also serves as an inhibitor of TLR signaling via binding to MyD88, which impairs its interaction with IRF5 and other downstream signaling elements.24 For example, Irf4-deficient mice develop an exaggerated postischemic inflammatory response aggravating ischemic acute renal failure, which depends on the oxidative stress-driven induction of IRF4 in intrarenal DCs.25 As another example, bacterial products specifically induce IRF4 in DCs of the intestinal wall, a mechanism that protects mice from experimental colitis.26 Hence, IRF4 suppresses innate immunity but fosters adaptive immunity. These amFigure 1. Lack of IRF4 increases the activation of antigen-presenting cells. (A) Serum bivalent immunoregulatory roles are again cytokine levels were determined by ELISA in B6lpr mice (black bars) and B6lpr/Irf4⫺/⫺ unique among the members of the IRF mice (white bars) at 6 months of age. (B) Spleen monocytes were stimulated with 1 family. ␮g/ml LPS and the cytokine levels were determined by ELISA. (C-E) Spleen cells were Given the clear roles of IRF1, IRF3, quantified by flow cytometry using surface activation markers as indicated. Data IRF5, and IRF7 in the pathogenesis of SLE represent mean ⫾ SEM from 12 to 14 mice in each group. *P ⬍ 0.05, **P ⬍ 0.01 versus and lupus nephritis and the multiple roles B6lpr mice. 1444

Journal of the American Society of Nephrology

J Am Soc Nephrol 22: 1443–1452, 2011

www.jasn.org

RESULTS IRF4 Deficiency Increases Serum Cytokine Levels by Activating DCs and Macrophages in B6lpr Mice

We generated Irf4-deficient B6lpr mice by crossing Irf4-deficient with Fas-deficient (lpr) C57BL/6 mice. Litters of B6lpr/Irf4⫺/⫺ mice were bred along Mendelian ratios and revealed no differences in body weight gain as compared with B6lpr (Irf4 wild-type) mice (not shown). In phenotyping both strains at 6 months of age, we first determined serum levels of various proinflammatory cytokines by ELISA. B6lpr/Irf4⫺/⫺ mice revealed significantly higher serum levels of TNF and IL-12p40 than their agematched B6lpr counterparts (Figure 1A), suggesting that IRF4 suppresses systemic cytokine release in B6lpr mice. Because IRF4 has been described to suppress the activation of antigen-

BASIC RESEARCH

presenting cells, we isolated splenocytes from B6lpr and B6lpr/Irf4⫺/⫺ mice and assessed cytokine production 24 hours after LPS stimulation in vitro. In fact, splenocytes from B6lpr/Irf4⫺/⫺ mice produced much higher levels of IFN␥, IL-12p40, TNF, and MCP-1/CCL2 as compared with cells from B6lpr mice (Figure 1B). Next we determined the activation state of various DC subsets in spleens of 6-month-old mice by flow cytometry. IRF4 deficiency increased the total numbers of CD11c/CD4-positive DCs and CD4/CD8 double-negative CD11c DCs (Figure 1C). IRF4 was required to suppress DC activation because B6lpr/Irf4⫺/⫺ mice displayed higher numbers of CD40-positive CD11c DCs and MHCII-positive F4/80/CD11c DCs (Figure 1D). In addition, lack of IRF4 increased the numbers of CD11b/F4/80-positive macrophages in spleens; most of them were MHCII positive, indicating a classically activated (M1) macrophage phenotype (Figure 1E). Alternatively activated CD206-positive macrophages remained in a minor splenocyte population in 6-month-old B6lpr/Irf4⫺/⫺ mice (Figure 1E). Taken together, lack of IRF4 increases systemic cytokine production and the activation of antigen-presenting cells in B6lpr mice, indicating an immunosuppressive role for IRF4 in these aspects of innate immunity. Lack of IRF4 Prevents Lupus Nephritis in B6lpr Mice

Next we questioned whether the aggravated systemic inflammation in B6lpr/Irf4⫺/⫺ mice is associated with an aggravation of lupus nephritis. At 6 months of age, B6lpr mice developed diffuse proliferative glomerulonephritis that was associated with diffuse mesangial matrix expansion, mesangial cell proliferation, and occasional tuft adhesions (Figure 2). Tuft necrosis and crescent formation were absent. On immunostaining, diffuse proliferative glomerulonephritis was associated with extensive glomerular IgM and IgG deposits presenting in mesangial (IgG) and capillary (IgM/IgG) staining patterns (Figure 2).

Figure 2. Lack of IRF4 abrogates renal pathology in 6-month-old B6lpr mice. Renal sections from 6-month-old B6lpr and B6lpr/Irf4⫺/⫺ mice were stained with periodic acid–Schiff (PAS), anti-mIgM, anti-mIgG, or anti-complement factor C9 as indicated. Note that B6lpr but not B6lpr/Irf4⫺/⫺ mice develop diffuse proliferative glomerulonephritis with IgM, IgG, and C9 deposits. Original magnification, ⫻400. J Am Soc Nephrol 22: 1443–1452, 2011

Figure 3. Lack of IRF4 abrogates lupus nephritis in 6-month-old B6lpr mice. The indices of lupus nephritis disease activity and chronicity were assessed by semiquantitative morphometry on PAS-stained renal sections from 6-month-old mice of both mouse strains as described in Concise Methods. Plasma creatinine levels were determined at the same age. Data represent mean ⫾ SEM from at least 12 mice per group. IRF4 in Lupus Nephritis

1445

BASIC RESEARCH

www.jasn.org

Staining for complement factor 9 showed mainly mesangial positivity (Figure 2), altogether indicating diffuse proliferative lupus-like immune complex glomerulonephritis as the renal manifestation of spontaneous autoimmunity in B6lpr mice. Lack of IRF4 completely abrogated this phenotype because age-matched B6lpr/Irf4⫺/⫺ mice developed hardly any glomerular Ig and complement deposits or glomerular abnormalities as shown by light microscopy (Figure 2). This was also evident by morphometrical assessment of the activity and chronicity index for lupus nephritis (Figure 3). Some renal sections showed a slight increase in activated mesangial cells in B6lpr/Irf4⫺/⫺ mice, but this was not a consistent finding. Abnormalities of the vascular or tubulointerstitial compartment were absent in both mouse strains. Because of the moderate renal lesions, plasma creatinine levels were only mildly elevated in B6lpr mice and there was a nonsignificant trend toward lower levels in B6lpr/Irf4⫺/⫺ mice (Figure 3). Albuminuria was absent in both strains (not shown). The abrogated lupus nephritis phenotype in B6lpr/Irf4⫺/⫺ mice was associated with lower mRNA expression levels of the proinflammatory chemokines CCL2, CCL5, CXCL2, and CXCL10 (Figure 4A), which correlated with a significant reduction of Mac2-positive glomerular macrophages (Figure 4B). Together, lack of IRF4 protects B6lpr mice from lupus nephritis. Lack of IRF4 Prevents Autoimmune Lung Disease in B6lpr Mice

Figure 4. Lack of IRF4 reduces renal chemokine mRNA expression in 6-month-old B6lpr mice. (A) Renal mRNA was isolated from 6-month-old B6lpr mice (black bars) and B6lpr/Irf4⫺/⫺ mice (white bars) and quantified by real-time reverse-transcriptase PCR. Data are expressed as mean of the ratio versus the respective 18S rRNA level ⫾ SEM. (B) Mac2-positive macrophages were quantified in 20 glomeruli per section in both mouse strains at 6 months of age. Data represent mean ⫾ SEM from at least 12 mice per group. *P ⬍ 0.05 versus B6lprmice.

Figure 5. Lack of IRF4 abrogates lung disease in 6-month-old B6lpr mice. (A) Lung sections from 6-month-old B6lpr and B6lpr/Irf4⫺/⫺ mice were stained with PAS. Note that B6lpr but not B6lpr/Irf4⫺/⫺ mice develop peribronchial and perivascular immune cell infitrates. Original magnification, ⫻100. (B) Mac2-positive glomerular cells were quantified in 20 glomeruli per section. Data are mean cell counts per glomerulus ⫾ SEM of 9 to 12 mice in each group. ***P ⬍ 0.001 versus B6lpr mice.

Autoimmune lung disease is another manifestation of SLE. At 6 months of age, B6lpr mice displayed focal areas of peribronchial and perivascular lymphocyte infiltrates (Figure 5). Such infiltrates were not detected in age-matched B6lpr/Irf4⫺/⫺ mice, indicating that lack of IRF4 protects B6lpr mice not only from lupus nephritis but also from autoimmune lung disease. IRF4 Is Required for the Production of Lupus Autoantibodies in B6lpr Mice

Lupus nephritis is a manifestation of immune complex disease in systemic autoimmunity. Glomerular immune complex deposits can develop in situ when circulating autoantibodies bind to nuclear particles that have deposited along the glomerular capillaries. Alternatively, circulating immune complexes get deposited along the glomerular filtration barrier.28 In any case, lupus autoantibodies represent an essential element in the 1446

Journal of the American Society of Nephrology

pathogenesis of lupus nephritis; therefore, we next examined the effect of the Irf4 genotype on lupus autoantibody formation. At the age of 6 months, B6lpr mice displayed significant hypergammaglobulinemia and antinuclear antibodies (ANAs) as well as autoantbodies specifically directed against doublestranded DNA (dsDNA) or the Smith antigen (Figure 6). By contrast, IRF4-deficient B6lpr mice had low serum IgG levels, and ANAs, dsDNA, and Smith autoantibodies were absent (Figure 6). Thus, IRF4 is required for the production of lupus autoantibodies in B6lpr mice. Lack of IRF4 Reduces Plasma Cells in B6lpr Mice

Serum IgG and pathogenic autoantibodies are derived from Ig-producing plasma cells. Given the low levels of circulating IgG and the absence of lupus autoantibodies in B6lpr/Irf4⫺/⫺ mice, we performed spleen cell flow cytometry for CD138 and J Am Soc Nephrol 22: 1443–1452, 2011

www.jasn.org

BASIC RESEARCH

of both genotypes (Figure 7B). We therefore conclude that lack of IRF4 abrogates lupus nephritis because B6lpr/Irf4⫺/⫺ mice are plasma cell deficient and no longer produce those autoantibodies that cause immune complex glomerulonephritis. Lack of IRF4 Reduces Th1 and Th17 Effector T Cells in B6lpr Mice

The various T cell subsets are essential regulators of autoimmune disease. Regulatory T cells suppress the expansion of autoreactive T cells and autoantigen-specific Th1 or Th17 effector T cells that promote inflammatory tissue injury.29,30 We therefore quantified T cell subsets in the spleens of 6-month-old B6lpr and B6lpr/Irf4⫺/⫺ mice by flow cytometry. The total number of CD3positive T cells was not affected by the Irf4 genotype, but CD8 T cells were slightly reduced and CD4/CD25/Foxp3-positive “regulatory” T cells were increased in B6lpr/Irf4⫺/⫺ versus B6lpr mice (Figure 8A). We identified Th1 and Th17 CD4 T cells by intracellular staining for IFN␥ or IL-17, respectively, and found that lack of IRF4 substantially reduced both of these subsets in B6lpr mice (Figure 8B). By contrast, IL-17 positivity in CD4/CD8 double-negative T cells was independent of the Irf4 genotype (not shown). Consistent with the latter finding, IL-17 and IL-21 were substantially reduced in the serum of 6-month-old B6lpr/Irf4⫺/⫺ mice (Figure 8C). Together, IRF4 deficiency impairs the maturation of Th1 and Th17 T cells as well as that of autoantibodyproducing plasma cells, which is associated with an abrogation of lupus nephritis in B6lpr mice. Figure 6. IRF4 deficient B6lpr mice lack hypergammaglobuminemia and autoantibody production. (A) B6lpr/Irf4⫺/⫺ (E) and B6lpr wild-type mice (F) were bled at the end of the study to determine serum levels of IgG, IgM, anti-Smith, and anti-dsDNA autoantibodies by ELISA. (B) ANAs were detected by staining of Hep2 cells using plasma dilutions of 1:40 as described in Concise Methods. Note the homogenous nuclear staining pattern using plasma from B6lprmice that was not detectable with plasma from B6lpr/Irf4⫺/⫺ mice.

␬ light chains to quantify plasma cells in 6-month-old B6lpr and B6lpr/Irf4⫺/⫺ mice. Lack of IRF4 was associated with a drastic reduction of the absolute numbers of spleen plasma cells on flow cytometry (Figure 7A). By contrast, the total numbers of mature B cells were not affected by the Irf4 genotype, although a shift from follicular- to marginal-zone B cells was observed in B6lpr/Irf4⫺/⫺ versus B6lpr mice (Figure 7A). The comparable numbers of mature B cells on flow cytometry were consistent with identical staining patterns for IgM-positive cells in mice J Am Soc Nephrol 22: 1443–1452, 2011

DISCUSSION

IRFs contribute to SLE and lupus nephritis in various ways. IRF1 acts as a nonredundant transcription factor that promotes the expression of many proinflammatory genes in immune and nonimmune cells.8,9 IRF3 and IRF7 mediate the expression of type I IFNs upon activation of innate viral nucleic acid sensors that, in lupus, can also be activated by endogenous nucleic acids and lupus autoantigens.7,12 IRF5 is required for immune cell maturation and for TLR signaling, two mechanisms that contribute to human SLE and lupus nephritis of Fc␥RIIB⫺/⫺Yaa or Fc␥RIIB⫺/⫺ mice.14 Here we show that IRF4 contributes to SLE and lupus nephritis in a different manner. On one side, IRF4 in Lupus Nephritis

1447

BASIC RESEARCH

www.jasn.org

tivation of DCs in postischemic kidneys, a process that limits local cytokine production and thereby prevents excessive renal pathology and acute renal failure.25 This immunosuppressive effect of IRF4 is related to its inducible expression in DCs, which blocks the interaction of IRF5 with the TLR adaptor MyD88 and thereby suppresses the local expression of NF␬B-dependent proinflammatory cytokines during bacterial or sterile forms of inflammation.24 Consistent with this concept, spleen DCs and macrophages in Irf4-deficient B6lpr mice were more activated and produced more proinflammatory cytokines as compared with their wild-type B6lpr counterparts. IRF4-deficient splenocytes also showed increased IFN-␥ production by natural killer (NK)1.1 cells, whereas IRF4 deficiency did not affect IFN-␥ producing T cells.31 This process should be sufficient to enhance autoimmunity and autoimmune tissue injury because enhanced antigen-presentation and co-stimulation is usually sufficient to increase the expansion of autoreactive B and T cells in lupus.32 For example, lack of the TLR signaling inhibitor single immunglobulin IL-1R-related molecule was similarly associated with activated DCs and increased production of lupus autoantibodies and subsequent immune complex glomerulonephritis in B6lpr mice.33 In addition, we recently observed the same for IL-1 receptorassociated kinase M, a factor that suppresses TLR signaling in DCs at the kinase level (unpublishedobservation).However,increasedactivation of antigen-presenting cells and massively increased serum cytokine levels were not associated with more autoimmunity or more Figure 7. IRF4 is required for plasma cells’ maturation. Flow cytometry was used to severe lupus nephritis in B6lpr/Irf4⫺/⫺ mice, determine the total number of distinct B and plasma cell subsets (A) in spleens of most likely because of the lack of autoimmunity 6-month-old B6lpr mice (black bars) and B6lpr/Irf4⫺/⫺ mice (white bars). The histogram and immune complex disease. presents mean ⫾ SEM of 12 to 14 mice in each group. *P ⬍ 0.05, ***P ⬍ 0.001 versus The lack of lupus-like autoimmunity as B6lpr mice. (B) IgM immunostaining of spleens identifies mature B cell distribution in indicated by the absence of ANAs and other mice of both genotypes. Original magnification, ⫻100. lupus autoantibodies was the most promiIRF4 suppresses innate immune recognition and therefore nent phenotype of B6lpr/Irf4⫺/⫺ mice. Our analysis identified an IRF4 deficiency enhances the activation of antigen-presenting almost complete absence of plasma cells, which was also illuscells including the production of NF␬B-dependent proinflamma- trated by hypogammaglobulinemia. IRF4 has a nonredundant tory cytokines. Although this process should enhance autoimmu- role in plasma cell maturation and Ig class switch recombinanity and autoimmune tissue inflammation, Irf4-deficient tion while germinal center B cell formation remains intact.19 As B6lpr mice still remain protected from glomerulonephritis such, Irf4-deficient mice are generally unable to induce hubecause IRF4 has a nonredundant role in the maturation of moral immune responses to antigen exposure,18 which obviplasma cells and Th1 and Th17 effector T cells, which pro- ously includes the production of ANAs in experimental lupus. tects B6lpr mice from the evolution of autoimmunity and These data document that innate immune activation is necesimmune complex disease. sary but not sufficient to cause lupus nephritis because autoanWe have recently shown that IRF4 potently suppresses the ac- tibody-mediated immune complex disease is an essential com1448

Journal of the American Society of Nephrology

J Am Soc Nephrol 22: 1443–1452, 2011

www.jasn.org

BASIC RESEARCH

cells,18,31 especially for T cell priming toward Th17 cells, because IRF4 phosphorylation by ROCK2 is required for the synthesis of IL-17 and of IL-21.21 As such, B6lpr/Irf4⫺/⫺ mice lack the major effector T cell populations that are involved in autoimmune tissue injuries including lupus nephritis beyond the production of lupus autoantibodies by plasma cells. In summary, IRF4 has nonredundant biologic effects for evolution of immune complex glomerulonephritis like the other members of the IRF family, namely IRF1, IRF3, IRF5, and IRF7. However, the mechanisms by which IRF4 contributes to autoimmune tissue injury differ from those of the other IRF family members. Although IRF4 deficiency activates antigen-presenting cells and induces systemic inflammation, lack of IRF4 also severely impairs plasma cell maturation and subsequent autoantibody production as well as the maturation of Th1 and Th17 effector T cells. As a consequence, IRF4 deficiency protects from the evolution of autoimmunity and lupus-like immune complex glomerulonephritis. It is therefore very likely that these immunoregulatory roles of IRF4 contribute to other lupus disease models in a similar manner. Together, we conclude that IRF4 is essential for the development of lupus-like autoimmunity and immune complex glomerulonephritis despite its suppressive effect on innate immunity.

CONCISE METHODS Animal Studies Irf4-deficient mice were generated, genotyped, and backcrossed to the C57BL/6J strain for ten generations as described previously.18 B6Irf4⫺/⫺ and B6lpr mice (Charles River) were mated to generate B6lpr/Irf4⫺/⫹ mice, which were then mated Figure 8. Lack of IRF4 impairs the maturation of Th17 cells. Flow cytometry was used to among each other to generate B6lpr/Irf4⫹/⫹ and determine (A) T cell subsets and IFN␥- and (B) IL-17-producing CD4 T cells in spleens of B6lpr/Irf4⫺/⫺ mice as described.33 Littermate fe6-month-old B6lpr mice (black bars) and B6lpr/Irf4⫺/⫺ mice (white bars). (C) Plasma IL-17 and males were used for all experimental procedures. IL-21 levels were determined by ELISA at 6 months of age. The histogram presents In each individual mouse, the genotype was asmean ⫾ SEM of 12 to 14 mice in each group. *P ⬍ 0.05, **P ⬍ 0.01 versus B6lpr mice. sured by PCR. Mice were housed in groups of five mice in sterile filter-top cages with a 12-hour dark/ ponent for the development of lupus nephritis. B6lpr/Irf4⫺/⫺ light cycle and unlimited access to autoclaved food and water. One cohort mice also lacked IFN␥-producing Th1 T cells and IL-17-pro- of mice was sacrificed at 24 weeks of age and one cohort was followed ducing Th17 T cells, both of which have been shown to con- until 52 weeks of age. All experimental procedures were performed actribute to glomerulonephritis30 and lupus nephritis in lpr mu- cording to the German animal care and ethics legislation and were aptant lupus mice.34,35 IRF4 is required for the maturation of T proved by the local governmental authorities. J Am Soc Nephrol 22: 1443–1452, 2011

IRF4 in Lupus Nephritis

1449

BASIC RESEARCH

www.jasn.org

Flow Cytometry Anti-mouse CD3, CD4, CD8, and CD25 antibodies (BD Pharmingen, Heidelberg, Germany) were used to detect CD3⫹CD4-CD8- doublenegative T cells and CD4⫹CD25⫹ regulatory T cell populations in spleens. Anti-CD11c was used to identify DCs, and the activation of CD11c-positive cells was assessed by co-staining for CD40 and MHCII (BD Pharmingen, Heidelberg, Germany). Anti-mouse B220, CD21, CD23, IgD, and IgM antibodies (BD Pharmingen) were used to detect mature B cells (B220⫹IgD⫹IgM⫹), marginal-zone B cells (B220⫹CD21highCD23low), and follicular B cells (B220⫹CD21lowCD23high). Spleen B1 cells were identified as CD19highB220lowCD5⫹CD43⫹ and peritoneal B1 cells were identified as CD19highB220lowCD5⫹. Anti-mouse CD19, CD5, and CD43 were procured from BD Biosciences. Plasma cells were identified using antimouse antibodies for Ig ␬ light chain and CD138 (BD Pharmingen). To identify the macrophage population in spleen, we used anti-mouse F4/80, CD11b, MHCII, and CD206 from BD Biosciences. Macrophages (F4/ 80⫹CD11b⫹), M1 macrophages (F4/80⫹CD11b⫹MHCII⫹), and M2 macrophages (F4/80⫹CD11b⫹CD206⫹) were identified as mentioned. Anti-mouse IL17a and IFN␥ (from BD Biosciences) were used along with anti-mouse CD3, CD4, CD8, and NK1.1 antibodies to evaluate Th1, TH17, and NK cells producing IFN␥. Respective isotype antibodies were used to demonstrate specific staining of cell subpopulations.36 Respective isotype antibodies were used to demonstrate specific staining of cell subpopulations. Quantification of cell number was done using counting beads for FACS (Invitrogen, Carlsbad, CA).

bodies, NUNC maxisorp ELISA plates were coated with poly-Llysine (Trevigen, Gaithersburg, MD) and mouse dsDNA. After incubation with mouse serum, dsDNA-specific IgG and serum IgG and serum IgM levels were detected by ELISA (Bethyl Laboratories, Montgomery, TX). For anti-Smith antibodies, NUNC maxisorp ELISA plates were coated with Smith antigen (Immunovision, Springdale, AR). A horseradish-peroxidase-conjugated goat anti-mouse IgG (Rockland, Gilbertsville, PA) was used for detection. Horseradish-peroxidase-conjugated anti-mouse IgG was used as the secondary antibody. Anti-nuclear antibodies were detected on Hep2 slides (1:50 diluted serum; BioRad Laboratories, Redmond, WA). Sections were mounted with Vectashield containing DAPI (Vector Laboratories, Burlingame, CA).

Real-Time Quantitative (TaqMan) Reverse-Transcriptase PCR Real-time reverse-transcriptase PCR was performed on mRNA from mouse organs as described previously.33 The SYBR Green Dye detection system was used for quantitative real-time PCR on a Light Cycler 480 (Roche, Mannheim, Germany). All of the technical steps were performed according to the Minimum Information for Publication of Quantitative Real-Time PCR Experiments guidelines.41 Controls consisting of double-deionized water were negative for target and housekeeper genes. Gene-specific primers (300 nM, Metabion, Martinsried, Germany) were designed and further analyzed in silico to target all known possible transcripts of interest. Primers are listed in Table 1.

Evaluation of Autoimmune Tissue Injury Lungs, spleens, livers, lymph nodes, and kidneys from all mice were fixed Statistical Analysis in 10% buffered formalin, processed, and embedded in paraffin. Sections One-way ANOVA followed by post hoc Bonferroni’s test was used (2 ␮m) for periodic acid–Schiff stains were prepared following routine for multiple comparisons using GraphPad Prism, version 4.03. Single groups were compared by unpaired two-tailed t test. Data protocols.37 The severity of the renal lesions was graded using the activity were expressed as mean ⫾ SEM. Statistical significance was asand chronicity indices for human lupus nephritis as described.38 sumed at P ⬍ 0.05. Autoimmune lung injury was scored semiquantitatively (0 to 4) by assessing the extent of peribronchial, perivascular, or interstitial lymphocyte infiltrates as described.39 For Table 1. Primers used for real-time RT-PCR IgM staining of the spleen sections, anti-mouse IgMAccession Gene Sequence ␮-chain specific antibodies (Vector, Burlingame, Number CA) were used. IRF4 NM013674 Forward primer: 5⬘-CAAAGCACAGAGTCACCTGG-3⬘ Complement component C9 antibody (kindly Reverse primer: 5⬘-TGCAAGCTCTTTGACACACA-3⬘ provided from Mohamed R. Daha, University of CCL2/MCP-1 NM011333 Forward primer: 5⬘-CCTGCTGTTCACAGTTGCC-3⬘ Reverse primer: 5⬘-ATTGGGATCATCTTGCTGGT-3´ Leiden, The Netherlands) was used at a 1:50 diluNM009140 Forward primer: 5⬘-CGGTCAAAAAGTTTGCCTTG-3⬘ tion, and secondary goat anti-rabbit biotinylated CXCL2/MIP2 Reverse primer: 5⬘-TCCAGGTCAGTTAGCCTTGC-3⬘ antibody (Vector, Burlingame, CA) was used at a NM013653 Forward primer: 5⬘-GTGCCCACGTCAAGGAGTAT-3⬘ 1:300 dilution. Serum cytokine levels were deter- CCL5 Reverse primer: 5⬘-CCACTTCTTCTCTGGGTTGG-3⬘ mined by ELISA following the manufacturer’s CXCL10 NM009140 Forward primer: 5⬘-ATGGATGGACAGCAGAGAGC-3⬘ protocols (IL-12p40: OptEiA, BD, Heidelberg, Reverse primer: 5⬘-GGCTGGTCACCTTTCAGAAG-3⬘ Germany; TNF: BioLegend, San Diego, CA; IL-4: IFN-␥ NM008337 Forward primer: 5⬘-ACAGCAAGGCGAAAAAGGAT-3⬘ OptEiA, BD, Heidelberg, Germany). Plasma creReverse primer: 5⬘-ACAGCAAGGCGAAAAAGGAT-3⬘ atinine levels were determined using a commercial TNF-␣ NM011609 Forward primer: 5⬘-CCACCACGCTCTTCTGTCTAC-3⬘ assay kit (DiaSys). Reverse primer: 5⬘-AGGGTCTGGGCCATAGAACT-3⬘

Autoantibody Analysis Serum antibody levels were determined by ELISA as described.40 For anti-dsDNA anti1450

IL-6

NM031168

18s RNA

NR003278

Journal of the American Society of Nephrology

Forward primer: 5⬘-TGATGCACTTGCAGAAAACA-3⬘ Reverse primer: 5⬘-ACCAGAGGAAATTTTCAATAGGC-3⬘ Forward primer: 5⬘-GCAATTATTCCCCATGAACG-3⬘ Reverse primer: 5⬘-AGGGCCTCACTAAACCATCC-3⬘

J Am Soc Nephrol 22: 1443–1452, 2011

www.jasn.org

ACKNOWLEDGMENTS The work was supported by a grant from the Deutsche Forschungsgemeinschaft (AN372/11-1 and GRK 1202) to H.J.A. The expert technical assistance of Dan Draganovic and Janina Mandelbaum is gratefully acknowledged. Parts of this work were performed as a medical thesis project by M.W. at the Medical Faculty of the University of Munich.

15.

16.

17.

DISCLOSURES

18.

None. 19.

REFERENCES 1. Kotzin BL: Systemic lupus erythematosus. Cell 85: 303–306, 1996 2. Goodnow CC: Multistep pathogenesis of autoimmune disease. Cell 130: 25–35, 2007 3. Gregersen PK, Olsson LM: Recent advances in the genetics of autoimmune disease. Annu Rev Immunol 27: 363–391, 2009 4. Kanta H, Mohan C: Three checkpoints in lupus development: Central tolerance in adaptive immunity, peripheral amplification by innate immunity and end-organ inflammation. Genes Immun 10: 390 –396, 2009 5. Theofilopoulos AN, Baccala R, Beutler B, Kono DH: Type I interferons (alpha/beta) in immunity and autoimmunity. Annu Rev Immunol 23: 307–336, 2005 6. Hom G, Graham RR, Modrek B, Taylor KE, Ortmann W, Garnier S, Lee AT, Chung SA, Ferreira RC, Pant PV, Ballinger DG, Kosoy R, Demirci FY, Kamboh MI, Kao AH, Tian C, Gunnarsson I, Bengtsson AA, RantapaaDahlqvist S, Petri M, Manzi S, Seldin MF, Ronnblom L, Syvanen AC, Criswell LA, Gregersen PK, Behrens TW: Association of systemic lupus erythematosus with C8orf13-BLK and ITGAM-ITGAX. N Engl J Med 358: 900 –909, 2008 7. Marshak-Rothstein A, Rifkin IR: Immunologically active autoantigens: The role of toll-like receptors in the development of chronic inflammatory disease. Annu Rev Immunol 25: 419 – 441, 2007 8. Wang Y, John R, Chen J, Richardson JA, Shelton JM, Bennett M, Zhou XJ, Nagami GT, Zhang Y, Wu QQ, Lu CY: IRF-1 promotes inflammation early after ischemic acute kidney injury. J Am Soc Nephrol 20: 1544 –1555, 2009 9. Reilly CM, Olgun S, Goodwin D, Gogal RM Jr, Santo A, Romesburg JW, Ahmed SA, Gilkeson GS: Interferon regulatory factor-1 gene deletion decreases glomerulonephritis in MRL/lpr mice. Eur J Immunol 36: 1296 –1308, 2006 10. Yasuda K, Richez C, Maciaszek JW, Agrawal N, Akira S, MarshakRothstein A, Rifkin IR: Murine dendritic cell type I IFN production induced by human IgG-RNA immune complexes is IFN regulatory factor (IRF)5 and IRF7 dependent and is required for IL-6 production. J Immunol 178: 6876 – 6885, 2007 11. Hoshino K, Sugiyama T, Matsumoto M, Tanaka T, Saito M, Hemmi H, Ohara O, Akira S, Kaisho T: IkappaB kinase-alpha is critical for interferon-alpha production induced by Toll-like receptors 7 and 9. Nature 440: 949 –953, 2006 12. Anders HJ: Pseudoviral immunity—A novel concept for lupus. Trends Mol Med 15: 553–561, 2009 13. Anders HJ, Krug A, Pawar RD: Molecular mimicry in innate immunity? The viral RNA recognition receptor TLR7 accelerates murine lupus. Eur J Immunol 38: 1795–1799, 2008 14. Richez C, Yasuda K, Bonegio RG, Watkins AA, Aprahamian T, Busto P, Richards RJ, Liu CL, Cheung R, Utz PJ, Marshak-Rothstein A, Rifkin IR:

J Am Soc Nephrol 22: 1443–1452, 2011

20.

21.

22.

23.

24.

25.

26.

27.

28. 29.

30.

31.

32.

BASIC RESEARCH

IFN regulatory factor 5 is required for disease development in the FcgammaRIIB⫺/⫺Yaa and FcgammaRIIB⫺/⫺ mouse models of systemic lupus erythematosus. J Immunol 184: 796 – 806, 2010 Savitsky DA, Yanai H, Tamura T, Taniguchi T, Honda K: Contribution of IRF5 in B cells to the development of murine SLE-like disease through its transcriptional control of the IgG2a locus. Proc Natl Acad Sci U S A 107: 10154 –10159, 2010 Tamura T, Yanai H, Savitsky D, Taniguchi T: The IRF family transcription factors in immunity and oncogenesis. Annu Rev Immunol 26: 535–584, 2008 Shaffer AL, Emre NC, Romesser PB, Staudt LM: IRF4: Immunity. Malignancy! Therapy? Clin Cancer Res 15: 2954 –2961, 2009 Mittrucker HW, Matsuyama T, Grossman A, Kundig TM, Potter J, Shahinian A, Wakeham A, Patterson B, Ohashi PS, Mak TW: Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 275: 540 –543, 1997 Klein U, Casola S, Cattoretti G, Shen Q, Lia M, Mo T, Ludwig T, Rajewsky K, Dalla-Favera R: Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol 7: 773–782, 2006 Zheng Y, Chaudhry A, Kas A, deRoos P, Kim JM, Chu TT, Corcoran L, Treuting P, Klein U, Rudensky AY: Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature 458: 351–356, 2009 Biswas PS, Gupta S, Chang E, Song L, Stirzaker RA, Liao JK, Bhagat G, Pernis AB: Phosphorylation of IRF4 by ROCK2 regulates IL-17 and IL-21 production and the development of autoimmunity in mice. J Clin Invest 120: 3280 –3295, 2010 Brustle A, Heink S, Huber M, Rosenplanter C, Stadelmann C, Yu P, Arpaia E, Mak TW, Kamradt T, Lohoff M: The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol 8: 958 –966, 2007 Satoh T, Takeuchi O, Vandenbon A, Yasuda K, Tanaka Y, Kumagai Y, Miyake T, Matsushita K, Okazaki T, Saitoh T, Honma K, Matsuyama T, Yui K, Tsujimura T, Standley DM, Nakanishi K, Nakai K, Akira, S: The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol 11: 936 –944, 2010 Negishi H, Ohba Y, Yanai H, Takaoka A, Honma K, Yui K, Matsuyama T, Taniguchi T, Honda K: Negative regulation of Toll-like-receptor signaling by IRF-4. Proc Natl Acad Sci U S A 102: 15989 –15994, 2005 Lassen S, Lech M, Rommele C, Mittruecker HW, Mak TW, Anders HJ: Ischemia reperfusion induces IFN regulatory factor 4 in renal dendritic cells, which suppresses postischemic inflammation and prevents acute renal failure. J Immunol 185: 1976 –1983, 2010 Watanabe T, Asano N, Murray PJ, Ozato K, Tailor P, Fuss IJ, Kitani A, Strober W: Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis. J Clin Invest 118: 545–559, 2008 Cohen PL, Eisenberg RA: Lpr and gld: Single gene models of systemic autoimmunity and lymphoproliferative disease. Annu Rev Immunol 9: 243–269, 1991 Glassock RJ: Human idiopathic membranous nephropathy—A mystery solved? N Engl J Med 361: 81– 83, 2009 Sakaguchi S, Miyara M, Costantino CM, Hafler DA: FOXP3⫹ regulatory T cells in the human immune system. Nat Rev Immunol 10: 490 –500, 2010 Summers SA, Steinmetz OM, Li M, Kausman JY, Semple T, Edgtton KL, Borza DB, Braley H, Holdsworth SR, Kitching AR: Th1 and Th17 cells induce proliferative glomerulonephritis. J Am Soc Nephrol 20: 2518 –2524, 2009 Lohoff M, Mittrucker HW, Prechtl S, Bischof S, Sommer F, Kock S, Ferrick DA, Duncan GS, Gessner A, Mak TW: Dysregulated T helper cell differentiation in the absence of interferon regulatory factor 4. Proc Natl Acad Sci U S A 99: 11808 –11812, 2002 Ohnmacht C, Pullner A, King SB, Drexler I, Meier S, Brocker T, Voehringer D: Constitutive ablation of dendritic cells breaks self-tolerance

IRF4 in Lupus Nephritis

1451

BASIC RESEARCH

33.

34.

35.

36.

37.

www.jasn.org

of CD4 T cells and results in spontaneous fatal autoimmunity. J Exp Med 206: 549 –559, 2009 Lech M, Kulkarni OP, Pfeiffer S, Savarese E, Krug A, Garlanda C, Mantovani A, Anders HJ: Tir8/Sigirr prevents murine lupus by suppressing the immunostimulatory effects of lupus autoantigens. J Exp Med 205: 1879 –1888, 2008 Balomenos D, Rumold R, Theofilopoulos AN: Interferon-gamma is required for lupus-like disease and lymphoaccumulation in MRL-lpr mice. J Clin Invest 101: 364 –371, 1998 Kyttaris VC, Zhang Z, Kuchroo VK, Oukka M, Tsokos GC: Cutting edge: IL-23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice. J Immunol 184: 4605– 4609, 2010 Kulkarni OP, Sayyed SG, Kantner C, Ryu M, Schnurr M, Sardy M, Leban J, Jankowsky R, Ammendola A, Doblhofer R, Anders HJ: 4SC-101, a novel small molecule dihydroorotate dehydrogenase inhibitor, suppresses systemic lupus erythematosus in MRL-(Fas)lpr mice. Am J Pathol 176: 2840 –2847, 2010 Kulkarni O, Pawar RD, Purschke W, Eulberg D, Selve N, Buchner K, Ninichuk V, Segerer S, Vielhauer V, Klussmann S, Anders HJ: Spiegel-

1452

Journal of the American Society of Nephrology

38.

39.

40.

41.

mer inhibition of CCL2/MCP-1 ameliorates lupus nephritis in MRL(Fas)lpr mice. J Am Soc Nephrol 18: 2350 –2358, 2007 Allam R, Pawar RD, Kulkarni OP, Hornung V, Hartmann G, Segerer S, Akira S, Endres S, Anders HJ: Viral 5⬘-triphosphate RNA and non-CpG DNA aggravate autoimmunity and lupus nephritis via distinct TLRindependent immune responses. Eur J Immunol, 38: 3487–3498, 2008 Sunderrajan EV, McKenzie WN, Lieske TR, Kavanaugh JL, Braun SR, Walker SE: Pulmonary inflammation in autoimmune MRL/Mp-lpr/lpr mice. Histopathology and bronchoalveolar lavage evaluation. Am J Pathol 124: 353–362, 1986 Patole PS, Pawar RD, Lichtnekert J, Lech M, Kulkarni OP, Ramanjaneyulu A, Segerer S, Anders HJ: Coactivation of Toll-like receptor-3 and -7 in immune complex glomerulonephritis. J Autoimmun 29: 52–59, 2007 Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, Mueller R, Nolan T, Pfaffl MW, Shipley GL, Vandesompele J, Wittwer CT: The MIQE guidelines: Minimum information for publication of quantitative real-time PCR experiments. Clin Chem 55: 611– 622, 2009

J Am Soc Nephrol 22: 1443–1452, 2011