the pathogenesis of lupus nephritis - nephrology now

10
BRIEF REVIEW www.jasn.org The Pathogenesis of Lupus Nephritis Maciej Lech and Hans-Joachim Anders Department of Nephrology, Medical Clinic and Polyclinic IV, University of Munich, Munich, Germany ABSTRACT Lupus nephritis is an immune complex GN that develops as a frequent complication of SLE. The pathogenesis of lupus nephritis involves a variety of pathogenic mechanisms. The extrarenal etiology of systemic lupus is based on multiple combinations of genetic variants that compromise those mechanisms normally assuring immune tolerance to nu- clear autoantigens. This loss of tolerance becomes clinically detectable by the presence of antinuclear antibodies. In addition, nucleic acids released from netting or apoptotic neu- trophils activate innate and adaptive immunity via viral nucleic acid-speci c Toll-like recep- tors. Therefore, many clinical manifestations of systemic lupus resemble those of viral infection. In lupus, endogenous nuclear particles trigger IFN-a signaling just like viral particles during viral infection. As such, dendritic cells, T helper cells, B cells, and plasma cells all contribute to the aberrant polyclonal autoimmunity. The intrarenal etiology of lupus nephritis involves antibody binding to multiple intrarenal autoantigens rather than the deposition of circulating immune complexes. Tertiary lymphoid tissue formation and local antibody production add to intrarenal complement activation as renal immunopa- thology progresses. Here we provide an update on the pathogenic mechanisms that lead to lupus nephritis and provide the rationale for the latest and novel treatment strategies. J Am Soc Nephrol 24: 13571366, 2013. doi: 10.1681/ASN.2013010026 SLE is a chronic autoimmune disease characterized by loss of tolerance against nuclear autoantigens, lymphoprolifera- tion, polyclonal autoantibody produc- tion, immune complex disease, and multiorgan tissue inammation. 1,2 SLE used to be referred to as a complex au- toimmune disease of unknown etiology; however, during the last decade, a multi- disciplinary approach to SLE research has built a more concise view of its path- ogenesis and for lupus nephritis (LN). Here we briey summarize an updated working model of SLE and LN, which provides a rationale for novel therapies. EXTRARENAL PATHOGENIC MECHANISMS OF LN Cell Death and Dead Cell Handling SLE develops from a loss of self-tolerance to ubiquitous nuclear autoantigens, which is a result of an immunization process. This observation implies two notions (Figure 1 and Table 1). First, au- toreactive, long-lived plasma cells, and memory T cells memorize their immu- nization against nuclei. These cells cannot be deleted by current immuno- suppressive therapies; hence, current treatments may suppress disease activity but do not cure SLE. 2,3 Second, the nu- clear antigens used for immunization had to be accessible to antigen-presenting cells, a process that is normally avoided by the homeostatic mechanism of rapid dead cell clearance. In fact, SLE develops in individuals with unfortunate combi- nations of genetic variants that, among other immunoregulatory defects, com- promise those mechanisms that nor- mally assure low levels of chromatin in extracellular compartments, particularly mutations that alter apoptosis, 4,5 the opsonization of dead cells by comple- ment, or their removal by phagocytes. 6 Neutrophils undergo NETosis, which re- leases nucleosomes into the extracellular extracellular space. 710 This nding re- cently revealed an unexpected role of neutrophils in SLE. 11 But how do dead cell clearance defects lead to SLE? Induction of Antiviral Immunity A delay of dead cell removal leads to degeneration of its components, which compromises those elements that nor- mally distinguish self-nucleic acids from viral nucleic acids. 12,13 For example, na- ture developed the methylation of DNA and RNA as a way to inhibit RNA and DNA recognition by Toll-like receptors (TLRs) 3, 7, and 9, a set of endosomal viral nucleic acid recognition receptors that trigger antiviral immunity during viral infection. 14 Therefore, in SLE pa- tients, nuclear particles are taken as viral particles that contain some protein com- ponent (antigen) as well as some immu- nostimulatory nucleic acid (immune adjuvant; Figure 1). During evolution, our immune sys- tem was primed to mount potent anti- viral immunity upon the recognition of viral particles, a response that is initiated against the components of virus-like nuclear particles in SLE patients. For Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. Hans-Joachim Anders, De- partment of Nephrology, Medical Clinic and Polyclinic IV, University of Munich, Ziemssenstr. 1, D-80336 Mu- nich, Germany. Email: [email protected] Copyright © 2013 by the American Society of Nephrology J Am Soc Nephrol 24: 13571366, 2013 ISSN : 1046-6673/2409-1357 1357

Upload: others

Post on 05-Oct-2021

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Pathogenesis of Lupus Nephritis - Nephrology Now

BRIEF REVIEW www.jasn.org

The Pathogenesis of Lupus Nephritis

Maciej Lech and Hans-Joachim Anders

Department of Nephrology, Medical Clinic and Polyclinic IV, University of Munich, Munich, Germany

ABSTRACTLupus nephritis is an immune complex GN that develops as a frequent complication ofSLE. The pathogenesis of lupus nephritis involves a variety of pathogenic mechanisms.The extrarenal etiology of systemic lupus is based on multiple combinations of geneticvariants that compromise those mechanisms normally assuring immune tolerance to nu-clear autoantigens. This lossof tolerancebecomesclinicallydetectableby thepresenceofantinuclear antibodies. In addition, nucleic acids released from netting or apoptotic neu-trophils activate innateandadaptive immunityvia viral nucleic acid-specificToll-like recep-tors. Therefore, many clinical manifestations of systemic lupus resemble those of viralinfection. In lupus, endogenous nuclear particles trigger IFN-a signaling just like viralparticles during viral infection. As such, dendritic cells, T helper cells, B cells, and plasmacells all contribute to the aberrant polyclonal autoimmunity. The intrarenal etiology oflupus nephritis involves antibody binding to multiple intrarenal autoantigens rather thanthe deposition of circulating immune complexes. Tertiary lymphoid tissue formation andlocal antibody production add to intrarenal complement activation as renal immunopa-thology progresses.Hereweprovide an update on thepathogenicmechanisms that leadto lupus nephritis and provide the rationale for the latest and novel treatment strategies.

J Am Soc Nephrol 24: 1357–1366, 2013. doi: 10.1681/ASN.2013010026

SLE is a chronic autoimmune diseasecharacterized by loss of tolerance againstnuclear autoantigens, lymphoprolifera-tion, polyclonal autoantibody produc-tion, immune complex disease, andmultiorgan tissue inflammation.1,2 SLEused to be referred to as a complex au-toimmune disease of unknown etiology;however, during the last decade, a multi-disciplinary approach to SLE researchhas built a more concise view of its path-ogenesis and for lupus nephritis (LN).Here we briefly summarize an updatedworking model of SLE and LN, whichprovides a rationale for novel therapies.

EXTRARENAL PATHOGENICMECHANISMS OF LN

Cell Death and Dead Cell HandlingSLE develops froma loss of self-toleranceto ubiquitous nuclear autoantigens,

which is a result of an immunizationprocess. This observation implies twonotions (Figure 1 and Table 1). First, au-toreactive, long-lived plasma cells, andmemory T cells memorize their immu-nization against nuclei. These cellscannot be deleted by current immuno-suppressive therapies; hence, currenttreatments may suppress disease activitybut do not cure SLE.2,3 Second, the nu-clear antigens used for immunizationhad to be accessible to antigen-presentingcells, a process that is normally avoidedby the homeostatic mechanism of rapiddead cell clearance. In fact, SLE developsin individuals with unfortunate combi-nations of genetic variants that, amongother immunoregulatory defects, com-promise those mechanisms that nor-mally assure low levels of chromatin inextracellular compartments, particularlymutations that alter apoptosis,4,5 the

opsonization of dead cells by comple-ment, or their removal by phagocytes.6

Neutrophils undergo NETosis, which re-leases nucleosomes into the extracellularextracellular space.7–10 This finding re-cently revealed an unexpected role ofneutrophils in SLE.11 But how do deadcell clearance defects lead to SLE?

Induction of Antiviral ImmunityA delay of dead cell removal leads todegeneration of its components, whichcompromises those elements that nor-mally distinguish self-nucleic acids fromviral nucleic acids.12,13 For example, na-ture developed the methylation of DNAand RNA as a way to inhibit RNA andDNA recognition by Toll-like receptors(TLRs) 3, 7, and 9, a set of endosomalviral nucleic acid recognition receptorsthat trigger antiviral immunity duringviral infection.14 Therefore, in SLE pa-tients, nuclear particles are taken as viralparticles that contain some protein com-ponent (antigen) as well as some immu-nostimulatory nucleic acid (immuneadjuvant; Figure 1).

During evolution, our immune sys-tem was primed to mount potent anti-viral immunity upon the recognition ofviral particles, a response that is initiatedagainst the components of virus-likenuclear particles in SLE patients. For

Published online ahead of print. Publication dateavailable at www.jasn.org.

Correspondence: Dr. Hans-Joachim Anders, De-partment of Nephrology, Medical Clinic and PolyclinicIV, University of Munich, Ziemssenstr. 1, D-80336 Mu-nich, Germany. Email: [email protected]

Copyright © 2013 by the American Society ofNephrology

J Am Soc Nephrol 24: 1357–1366, 2013 ISSN : 1046-6673/2409-1357 1357

Page 2: The Pathogenesis of Lupus Nephritis - Nephrology Now

example, ribonucleoprotein, U1snRNP,ligatesTLR7to induce type I IFNrelease inplasmacytoid dendritic cells,15 a processthat is tightly controlled by IL-1 receptor–associated kinase-M.16 RNA immunecomplexes activate B cells to produce an-tinuclear antibodies,17 which is con-trolled by the TIR8 gene encoding forthe SIGIRR protein.18,19 NucleosomalDNA or DNAwithin immune complexescan activate TLR9 on plasmacytoid den-dritic cells and drive B cell proliferation.20

Blockade of TLR7, TLR9, or both abro-gates type I IFN induction, SLE, and LNin mice.21–23 This (pseudo)antiviral im-mune response involves all antigen-pre-senting cells, particularly dendritic cellsand B cells, but only plasmacytoid den-dritic cells secrete large amounts of type IIFNs to set off an antiviral immuneresponse.5,24 The signature for IFNreceptor–dependent secondary gene ex-pression includes multiple antiviral andproliferative genes such as IFIT1, MX1,MX2, ISG15, and the OAS gene family,the IFN regulatory factors IRF7 andIRF5, as well as proinflammatory che-mokines CXCL10 and CXCL5, which al-together account for the nonspecificsymptoms shared by viral infectionsand SLE, such as fever, fatigue, arthralgia,and myalgia (Figure 2).25

Aberrant Lymphocyte ProliferationDendritic cells and B cells both have thecapacity to process antigens and presentantigens to T cells and they can substituteeach other for this purpose.26 Dendriticcells have a limited life span but their per-sistent activation by lupus autoantigensby TLR7 and TLR9 enhances their sur-vival and renders them resistant to gluco-corticoid-induced death.21 Persistentactivation of antigen-presenting cellsturns the interpretation of autoantigensfrom immune ignorance and lymphocyteanergy into lymphocyte activation andproliferation, which can overcome thefunctional unresponsiveness or anergyof mature autoreactive B cells.27 Murinedouble minute-2 is one of these mito-genic factors that is specifically inducedby DNA recognition.28 Murine doubleminute-2 neutralizes p53-dependent cellcycle arrest, which explains themitogenic

Figure 1. Pathomechanisms of LN outside the kidney. (A) Genetic variants of homeostaticcell death (i.e., Fas variants) and the rapid clearance of dead cell corpses (e.g., C3/4variants or DNAses variants) result either in secondary necrosis or incomplete chromatindigestion, which both promote the exposure of nuclear particles to the immune system.(B) Nuclear particles resemble viral particles and activate the same viral nucleic acidrecognition receptors on antigen-presenting cells. Genetic variants of those signalingelements are recognized to be risk factors for SLE. The activation of antigen-presentingcells changes (by costimulation) the immune interpretation of concomitantly presentedantigens of the same particle. (C) Polyclonal lymphocyte expansion hasmultiple effects onthe disease process and genetic variants further affect the differentiation of T helper cells.The complex regulation of lymphocyte activation and expansion is affected by multiplegenetic variants. The susceptibility genes and genes/molecules that are involved withineach biologic pathway are listed to the right: C1q, C2, C4A/B, C-reactive protein (CRP),a-glucoside transporter 5 (ATG5), three prime repair exonuclease 1 (TREX1), B cell CLL/lymphoma 2 (Bcl-2), IL-2 receptor a (IL-2Ra), tyrosine-protein kinase receptor 3 (TYRO3),mast/stem cell growth factor (MGF), Fcg receptor (FcGR), HLA IL-1 receptor–associatedkinase (HLA IRAK), IFN regulatory factor (IRF), signal transducer and activator of tran-scription (STAT), integrin aM (ITGAM), TNF a-induced protein 3 (TNFAIP3), zinc fingerprotein 36 (Zfp-36), IL-4, IFN-g, MHCI, MHCII, TNF, TLR7, single Ig and Toll-IL 1 receptor(SIGIRR), B cell scaffold protein with ankyrin repeats 1 (BANK1), B lymphoid tyrosine ki-nase (BLK), IKAROS family zinc finger 1 (IKZF1), protein tyrosine phosphatase, non-receptor type 22 (PTPN22), pituitary tumor-transforming 1 (PTTG1), RAS guanyl releasingprotein 3 (RASGRP3), TNF (ligand) superfamily, member 4 (TNFSF4), TNFAIP3-interactingprotein 1 (TNIP1), transmembrane activator and calcium-modulator and cyclophilin ligandinteractor (TACI), BAFF/BLyS, cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmedcell death protein 1 (PD-1/PDCD-1), and Gadd45 (activated by the stress-inducibleGADD45).

1358 Journal of the American Society of Nephrology J Am Soc Nephrol 24: 1357–1366, 2013

BRIEF REVIEW www.jasn.org

Page 3: The Pathogenesis of Lupus Nephritis - Nephrology Now

effect of endogenous DNA or DNA vi-ruses on autoreactive lymphocytes inSLE. Other lymphocyte mitogens are Blymphocyte stimulator (BlyS, also namedB cell activating factor [BAFF]) and aproliferation-inducing ligand (APRIL),which regulate B cell differentiation andIg class switching.29 The two APRIL/BLyS receptors transmembrane activatorand CAML-interactor (TACI) and B cellmaturation protein promote plasma cellsurvival; therefore, they are currentlyconsidered as potential therapeutic targetsin SLE.30

Furthermore, TNF-like weak inducerof apoptosis, a proinflammatory cyto-kine of the TNF superfamily, regulatesmultiple processes in autoimmunity andtissue inflammation through activationof the fibroblast growth factor–inducible14 receptor. B cell maturation towardautoantibody-producing plasma cellsinvolves IFN-related factor-4.31

Environmental Triggers of SLEActivityViral infections induce IFN-a release,which triggers antiviral immunity aswell as lupus disease activity.25 Bacterialinfections have a nonspecific immunos-timulatory effect, which involves atransient expansion of autoreactive lym-phocyte clones. Furthermore, bacterial

products stimulate intrarenal immunecells and renal cells, which can trigger atransient aggravation of proteinuria andkidney damage. Another environmentaltrigger of SLE activity is ultraviolet light,which induces an increase in the load ofdead cells by causing keratinocytedeath.32 In patients with a significantdead cell clearance defect, this processwill lead to increased levels of extracel-lular nuclear material, and additionalexposure of autoantigens and autoadju-vants to the immune system.1 Drug-induced SLE involves inhibition ofmethyl-transferases, a process that enhancesthe unmasking of endogenous nucleic acidsand the activation of TLR7 and TLR9.33,34

Progesterone and estrogens stimulate thesex hormone–dependent immunoregu-latory pathways.35

Together, SLE develops from a pecu-liar combination of genetic variants thatimpair those mechanisms that normallyprevent the exposure of nuclear particlesto the immune system and their capacityto activate viral recognition nucleic acidreceptors (Figure 1). Therefore, an IFN-a–dependent (pseudo)antiviral im-mune response accounts for thosenonspecific SLE symptoms that areshared with viral infections. The autoad-juvant activity of endogenous nucleicacids promotes an adaptive immune

response against the components of thenuclear particle, a process identical tovaccination. This implies the expansionof T and B cell clones with specificitiesfor predominantly nuclear autoantigensthat account for the production of anti-nuclear antibodies, immune complexdisease, and T cell–dependent tissuedamage. Hormonal and environmentalstimuli can enhance these processes atdifferent levels.

INTRARENAL PATHOGENICMECHANISMS OF LN

Immune Complex-Mediated RenalImmunopathologyThe nonspecific activation of autoreactiveB cells explains the polyclonal autoanti-body response leading to the diagnostichallmark of LN, the full house pattern ofIgM, IgA, and IgG deposits.36 However,antibody-deficient mice still develop LN;therefore, B cells have pathogenic effectsbeyond antibody production,37 includ-ing autoantigen presentation to activateautoreactive T cells and local proinflam-matory effects.26 Immune complexesdeposit in the mesangium or the suben-dothelial and subepithelial spaces or inperitubular capillaries depending on thequality of the autoantibodies, the dura-tion, and severity of LN.38 This impliesthat immune complex formation in themesangium causes class I and II lesions,subendothelial immune complex forma-tion in class III and IV lesions, and sub-epithelial immune complexes in class Vlesions as well as the overlapping formsIII/IV and IV/V.39 The traditional con-cept that circulating immune complexesin lupus passively deposit in the kidneyhas been challenged by novel data.40,41

Glomerular immune complexes ratherform in situ by secondary binding to nu-cleosomes from renal cells.42 Anotherpotential intrarenal source of nucleo-somes are neutrophils upon NETosis,11

due to the release of neutrophil extracel-lular traps that is initiated by anti-LL37antibodies.7–10 Heparin modulates theintrarenal effect of chromatin either byenhancing the DNA-I–dependent chro-matin degradation or by preventing

Table 1. Pathomechanisms of LN inside the kidney

Glomerular Pathology Tubulointerstitial Pathology

Mesangial and subendothelial, immunecomplex deposits, complement activation

Immune complex deposits inperiglomerular vessels

Fc, Toll-like, and complement receptor activation Complement activationActivation of renal cells and infiltrating leukocytes(subepithelial IC causes LN class V andpodocyte injury with massive proteinuria)

Activation of endothelial cells, luminaladhesion molecules

Local cytokine expression Leukocyte recruitmentRecruitment of leukocytes Local antibody production by B cells including

tertiary lymphoid organ formationProliferation of endothelial and mesangial cells Cytotoxic and Th17 T cellsFiltration barrier damage causing proteinuriaand hematuria

Proapoptotic cytokines

Renal cell necrosis causing focal scaring Proximal tubular cell damage causingproteinuria

Proliferation of parietal epithelial cells andcrescent formation

Tubular/vascular atrophy

Periglomerular inflammation Hypoxia → inflammationGlobal glomerulosclerosis Insufficient tubular and vascular repair plus

ischemia promotes interstitial fibrosis

J Am Soc Nephrol 24: 1357–1366, 2013 Pathogenesis of Lupus Nephritis 1359

www.jasn.org BRIEF REVIEW

Page 4: The Pathogenesis of Lupus Nephritis - Nephrology Now

chromatin binding to the glomerularbasement membrane.43 Anti-DNA anti-bodies activate endothelial and mesangialcells through different mechanisms. Forexample, antibodies are directly taken upinside renal cells.41 This process involvescross-reactivity with a-actinin or annexinII on mesangial cells,44,45 but this conceptcould not be confirmed by recent stud-ies.46,47 In addition, intrarenal immune

complex deposits activate comple-ment,36 which demonstrates the dualrole of complement factors in LN.48Com-plement deficiency impairs opsonizationand removal of lupus autoantigens fromthe extracellular space, whereas comple-ment factors also directly cause immunecomplex–related renal inflammation andimmunopathology.49,50 That immunecomplexes activate glomerular cells by

Fc receptor (FcR) ligation is also well es-tablished even though the data accumu-lated over the last few decades remaincomplex.51 Subepithelial immune com-plex deposits lead to secondary membra-nous GN and nephrotic syndrome bydamaging podocytes.

Intrarenal Activation of TLRs andIFN SignalingThe nucleic acid component of immunecomplexes also activates intrarenal in-flammation by TLRs in intrarenal mac-rophages and dendritic cells.52 Inaddition, immunostimulatory nucleicacids activate glomerular endothelium,mesangial cells, and macrophages toproduce large amounts of proinflamma-tory cytokines and IFN-a and IFN-b.53–59

The functional significance of thisintraglomerular IFN signaling is poorlyunderstood but seems to contribute torenal damage in LN and should triggerthe formation of tubuloreticular struc-tures or inclusions that represent anultrastructural characteristic of IFNsignaling.56,60 Together, the ligation ofTLRs, complement receptors, and FcRsactivates renal cells to release proinflam-matory cytokines and chemokines, andinduces the luminal expression of selectinsand adhesion molecules inside the micro-vasculature.61,62

Chemokine-Mediated Recruitmentof Different Leukocyte SubsetsCytotoxicTcells, Th17Tcells, aswell asBcells infiltrate the kidney in LN.24 Themembers of the chemokine family spe-cifically direct different leukocyte sub-sets by distinct chemokine receptorsinto different renal compartments.63

For example, the chemokine CCL2 re-cruits CCR2+ proinflammatory macro-phages and T cells into the glomerulusand the tubulointerstitium,64,65 whereasCCR1+ cells only recruit to the intersti-tial compartment and not to the glomer-ulus in LN.66 Other leukocyte subsetsinvolve other chemokines and chemo-kine receptors for their recruitmentinto the kidney.63,67 Leukocyte recruit-ment is tightly regulated. For example,renal cells produce pentraxin-3, whichhas the potential to directly inhibit

Figure2. Therapeutic targets for LN.Aberrant immunity in SLE involvesmanydifferent celltypes and cytokine mediators, which could be suitable therapeutic targets. MCP-1,monocyte chemotactic protein-1; CCR, CC chemokine receptor; CXCR, C-X-C chemokinereceptor.

1360 Journal of the American Society of Nephrology J Am Soc Nephrol 24: 1357–1366, 2013

BRIEF REVIEW www.jasn.org

Page 5: The Pathogenesis of Lupus Nephritis - Nephrology Now

leukocyte recruitment by interferingwith P-selectin on the surface of acti-vated endothelial cells.68,69 In lupus-like autoimmunity of C57BL/6lpr/lpr

mice, the role of pentraxin-3 seems tobe organ specific, because it suppresseslymphocyte recruitment to the lungs butnot to the kidney.70 Infiltrating leuko-cytes form de novo perivascular tertiarylymphoid organs inside the kidney,which involve the clonal expansion andongoing somatic hypermutation of Bcells in proximity to T cell aggregates.71

Such B cells undergo intrarenal prolifera-tion and activation, which contributes tolocal inflammation and tissue pathologyin addition to their role for systemic andintrarenal autoantibody production.26,72,73

These data provide a rationale for B cell–targeted therapies in LN.74,75 T cellinfiltrates also contribute to immunopa-thology in LN, particularly IL-17 produc-ing CD3+/CD4+ or CD3+CD4/82/2

T cells.76 Macrophages also contributeto renal damage, particularly F4/80(hi)/CD11c(int)Gr1(lo)/Ly6C(lo)/VLA4(lo)/MHCII(hi)/CD43(lo)/CD62L(lo) mac-rophages.77,78

Maladaptive Tissue RepairContributes to CKD ProgressionDamage to renal parenchymal cells trig-gers healing responses that contribute torenal pathology. Focal tuft necrosis isfollowed by a migration of parietal epi-thelial cells in the glomerular tuft, wherethey produce extracellular matrix con-tributing to FSGS progressing to globalglomerulosclerosis.79 During this pro-cess, the parietal cells maintain their po-larized epithelial phenotype and laydown extracellular matrix on top of thepodocytes.79 In addition, cellular glo-merular crescent formation resultsfrom activation of parietal epithelial cellsthat fill the urinary space by uncoordi-nated proliferation.80,81 This process canbe triggered by glomerular basementmembrane breaks that allow plasmaleakage into Bowman’s space, where mi-togenic plasma components such as fi-brinogen trigger hyperproliferation ofthe parietal epithelial cells.82 At laterstages, the parietal epithelial cells losetheir polarity and produce matrix all

around themselves, which creates hon-eycomb matrix deposits in Bowman’sspace that turn cellular crescents into fi-brocellular crescents with glomerulo-sclerosis, also referred to as class VI LN.

THE PRESENT AND FUTURE OF LNTHERAPY

Nonselective ImmunosuppressantsSteroids, cyclophosphamide, azathio-prine, andmycophenolate mofetil remainfirst-line therapeutics for treatment ofLN. These nonselective immunosuppres-sants have much improved the responserates of acute manifestations and theoverall mortality of SLE; however, thelong-term outcomes of LN have notfurther improved during the last 30years.83 High-dose steroids and cyclo-phosphamide are frequently associatedwith severe side effects, and infectionscontribute to the overall mortality inSLE. The same applies to the other non-selective immunosuppressants. Forexample, in the ALMS (Apreva LupusManagement) trial, infection-relatedmortality was even higher in patientstreated with mycophenolate mofetilthan in the group treated with high-dose cyclophosphamide. Reducing thedrug dose was the first strategy to limitside effects, and some researchers won-der whether oral cyclophosphamidetherapy is no longer needed.84 In addi-tion, some studies suggest that Caucasianpatients may no longer need high-dosecyclophosphamide, because the Euro-Lupus trial demonstrated favorablelong-term outcomes with much lowerdoses of cyclophosphamide.85 Otherimmunosuppressants like dihydrooro-tate dehydrogenase inhibitors add tothe current choices but are still nonspe-cific.86,87 However, lupus drug develop-ers continue to search for new drugsthat more specifically modulate the ab-errant immunity in SLE with fewer sideeffects.

Novel Moieties that Target SpecificLeukocyte SubsetsOne of the current strategies to specificallyinterfere with systemic autoimmunity in

SLE is to use cell type–specific drugs. Forexample, great hope was put into thestrategy of B cell–directed therapy as Bcells are the source of autoantibody pro-duction.88 Meanwhile, it is clear that Bcells in SLE also contribute to autoimmu-nity and tissue inflammation in manyother ways, which increases the hopethat depleting or modulating B cellswould result in major benefits in SLEand LN.89 This led to the developmentof the fully humanized anti-CD20 anti-body (rituximab), the anti-CD22 anti-body (epratuzumab), and to anti-BlyS(belimumab). Because the randomizedplacebo-controlled Lupus Nephritis As-sessment with Rituximab trial failed todemonstrate a benefit of add-on rituximabfor the induction therapy of incident LNclass III/IV/V, this approach to B cell de-pletion still has questions.90,91 At about thesame time, the Exploratory Phase II/IIISLE Evaluation of Rituximab trial alsofailed to demonstrate benefits on nonrenallupus.91 However, uncontrolled studies onrefractory LN still document 75% re-sponder rates and many specialists con-tinue using rituximab successfully forthese patients.92

BLyS blockade is another promisingstrategy to target B cell proliferation. Alarge randomizedplacebo-controlled tri-al suggested that add-on belimumab ontop of standardmaintenance therapy cansignificantly improve persistent diseaseactivity up to 72 weeks.93 This study ledto the US Food and Drug Administra-tion and European Medicines Agencyapproval of belimumab for nonrenal lu-pus in the United States and Europe. Pa-tients with severe LN were excludedfrom the BLISS-56 and BLISS-76 trials,but data from those patients with mod-erate nephritis raise hope that belimumabcould also be efficient in severe LN.94

Such a trial is currently underway. OtherB cell–directed strategies include atacicept(TACI-Ig), LY2127399 (anti-BAFF), andanti-BR3 (anti-BAFF-R).89

Dendritic cell–T cell interaction is atarget of costimulatory ligand/receptorblockers such as CTLA-4-Ig (abatacept).This drug blocks the interaction of CD80and CD86 on antigen-presenting cellswith CD28 on T cells, which suppresses

J Am Soc Nephrol 24: 1357–1366, 2013 Pathogenesis of Lupus Nephritis 1361

www.jasn.org BRIEF REVIEW

Page 6: The Pathogenesis of Lupus Nephritis - Nephrology Now

T cell activation.95 Abatacept suppressedlupus in mice but did not prevent flaresin SLE patients.96 Three trials with anti-CD40L failed to demonstrate efficacy.Abetimus is a drug that modulates auto-immunity by altering antigen recogni-tion by T cells. Abetimus is composedof a series of linked oligonucleotides,which block the binding of anti-dsDNAantibodies to their autoimmune targetsand tolerize B cells with antigen specific-ity for DNA. Unfortunately, the resultsin clinical trials have been very modest.A similar approach is followed byedratide, a peptide derived from theantigen-binding region of a humanmonoclonal anti-dsDNA antibody. It hasbeen proposed that this molecule canmodulate the function of DNA-reactiveB cells through idiotype–anti-idiotype in-teractions but again, the convincing dataon efficacy are still lacking.

The concept of anti-dsDNA–specifictherapeutic interventions is no longerpreferred because it targets only a verysmall subset of B cells, and no longerseems sufficient in view of the failing Bcell depletion trials and the fact thatdsDNA antibodies certainly contributeto SLE but remain only one of many dif-ferent pathogenic elements. Finally,plasma cells now appear as an attractivetherapeutic target in SLE because theyharbor the long-term memory of hu-moral immunity and produce the lupusautoantibodies.97 Rituximab does notdeplete plasma cells, because these arenegative for CD20.97 Massive antibodyproduction in plasma cells involves theintracellular proteasome complex forprotein processing. The proteasome in-hibitor bortezomib was proven to be ef-fective in mouse models of LN,98,99 butclinical trials with bortezomib in humanLN is still pending.

Additional Innovative TherapeuticStrategiesThe pseudo-antiviral immunity conceptis based on the molecular mimicry ofendogenous nucleic acids at the viral nu-cleic acid recognition receptors TLR7 andTLR9.100 Hence, blocking these TLRs andthe subsequent IFN signaling are additiveto established therapeutic targets in

SLE.101 Current treatment guidelinesrecommend hydroxychloroquine treat-ment for all SLE patients, including allpatients with LN.102–104 Antimalarialdrugs like hydroxychloroquine inhibitlysosomal acidification, which blocksthe adjuvant effect of endogenous nu-cleic acids by TLR7 and TLR9 duringthe lysosomal processing of nuclear par-ticles in endolysosomal compartmentsof antigen-presenting cells.20 Mean-while, more specific TLR7 and TLR9agents have been developed that effec-tively suppressed LN in murine SLEmodels and are now being tested in clin-ical trials.21–23,105 Antagonism of IFN-ais feasible with IFN-a antibodies. Adouble-blind randomized study withsifalimumab, a fully human anti-IFN-amAb, demonstrated that IFNa drivesthe overexpression of IFN-dependentgenes in human SLE, which is reversedby sifalimumab.106 Other cytokine-directed innovative therapies includeanti-Tweak (ATLAS trial) as well as anti-IL6R (tocilizumab). The current status foroff-label use of these drugs was recentlysummarized.30,107 Finally, it remains an at-tractive notion to add anti-inflammatoryagents to immunosuppressive drugs to re-duce the degree of therapeutic immuno-suppression and the risk of therapy-relatedinfections. As a proof of concept, the com-bination therapy of the CCL2 chemokineantagonistic Spiegelmer,mNOX-E36, and25% of full-dose cyclophosphamide wasshown to be as effective as 100% cyclophos-phamide to control severe SLE in MRLlpr/lprmice108; thus, the cyclophosphamide-related T cell ablation and myelosuppres-sion couldbeprevented,whilemaintainingtreatment efficacy.108

SUMMARY

The pathogenesis of LN involves extrare-nal and intrarenalpathogenicmechanisms.The extrarenal factors include complexcombinations of genetic variants thatare different in each patient, which ex-plains the variability of clinical mani-festations. SLE develops when geneticvariants compromise those mechanismsthat normally assure immune tolerance

for nuclear autoantigens. Loss of toler-ance becomes clinically evident by thepresence of antinuclear antibodies. Thenucleic acid content of nuclear particlesfrom netting or apoptotic neutrophilsactivates innate and adaptive immunityby TLR7 and TLR9, which triggers anIFN-a–mediated antiviral host defenseprogram that accounts for many of thenonspecific SLE symptoms. As such,dendritic cells, T helper cells, B cells,and plasma cells all contribute to the ab-errant polyclonal autoimmunity. The in-trarenal etiology of LN involves antibodybinding to intrarenal nuclear autoanti-gens, local complement, and FcR activa-tion. Tertiary lymph follicles, to somedegree, form inside the kidney, which in-clude B cells with local proinflammatoryeffects as well as plasma cells that secreteautoantibody inside the kidney. These in-sights into the pathogenesis of lupus pro-vide the rationale for a number of noveltherapeutic targets.

ACKNOWLEDGMENTS

This work was supported by the German

Research Foundation (grants AN372/11-1

and GRK 1202).

DISCLOSURES.None.

REFERENCES

1. Liu Z, Davidson A: Taming lupus-a new un-derstanding of pathogenesis is leading toclinical advances. Nat Med 18: 871–882,2012

2. Tsokos GC: Systemic lupus erythematosus.N Engl J Med 365: 2110–2121, 2011

3. Goodnow CC: Multistep pathogenesis ofautoimmune disease.Cell 130: 25–35, 2007

4. Saxena R, Mahajan T, Mohan C: Lupus ne-phritis: Current update. Arthritis Res Ther13: 240, 2011

5. Migliorini A, Anders HJ: A novel pathoge-netic concept-antiviral immunity in lupusnephritis. Nat Rev Nephrol 8: 183–189,2012

6. Muñoz LE, Lauber K, Schiller M, ManfrediAA, Herrmann M: The role of defectiveclearance of apoptotic cells in systemic au-toimmunity. Nat Rev Rheumatol 6: 280–289, 2010

1362 Journal of the American Society of Nephrology J Am Soc Nephrol 24: 1357–1366, 2013

BRIEF REVIEW www.jasn.org

Page 7: The Pathogenesis of Lupus Nephritis - Nephrology Now

7. Hakkim A, Fürnrohr BG, Amann K, Laube B,Abed UA, Brinkmann V, Herrmann M, VollRE, Zychlinsky A: Impairment of neutrophilextracellular trap degradation is associatedwith lupus nephritis. Proc Natl Acad Sci U SA 107: 9813–9818, 2010

8. Villanueva E, Yalavarthi S, Berthier CC,Hodgin JB, Khandpur R, Lin AM, Rubin CJ,Zhao W, Olsen SH, Klinker M, Shealy D,Denny MF, Plumas J, Chaperot L, KretzlerM, Bruce AT, KaplanMJ: Netting neutrophilsinduce endothelial damage, infiltrate tissues,and expose immunostimulatory molecules insystemic lupus erythematosus. J Immunol187: 538–552, 2011

9. Garcia-Romo GS, Caielli S, Vega B,Connolly J, Allantaz F, Xu Z, Punaro M,Baisch J, Guiducci C, Coffman RL, Barrat FJ,Banchereau J, Pascual V:Netting neutrophilsaremajor inducers of type I IFNproduction inpediatric systemic lupus erythematosus. SciTransl Med 3: 73ra20, 2011

10. Lande R, Ganguly D, Facchinetti V, Frasca L,ConradC,Gregorio J,Meller S, ChamilosG,Sebasigari R, Riccieri V, Bassett R, Amuro H,Fukuhara S, Ito T, Liu YJ, Gilliet M: Neu-trophils activate plasmacytoid dendriticcells by releasing self-DNA-peptide com-plexes in systemic lupus erythematosus. SciTransl Med 3: 73ra19, 2011

11. Bosch X: Systemic lupus erythematosus andthe neutrophil.NEngl JMed 365: 758–760,2011

12. Hof D, Raats JM, Pruijn GJ: Apoptotic mod-ifications affect the autoreactivity of the U1snRNP autoantigen.Autoimmun Rev 4: 380–388, 2005

13. Huck S, Deveaud E, Namane A, Zouali M: Ab-normal DNA methylation and deoxycytosine-deoxyguanine content in nucleosomes fromlymphocytes undergoing apoptosis. FASEBJ 13: 1415–1422, 1999

14. Karikó K, Ni H, Capodici J, Lamphier M,Weissman D: mRNA is an endogenous li-gand for Toll-like receptor 3. J Biol Chem279: 12542–12550, 2004

15. Savarese E, Chae OW, Trowitzsch S, WeberG, Kastner B, Akira S, Wagner H, SchmidRM, Bauer S, Krug A: U1 small nuclear ribo-nucleoprotein immune complexes inducetype I interferon in plasmacytoid dendriticcells through TLR7. Blood 107: 3229–3234,2006

16. Savarese E, Steinberg C, Pawar RD, ReindlW, Akira S, Anders HJ, Krug A: Requirementof Toll-like receptor 7 for pristane-inducedproduction of autoantibodies and develop-mentofmurine lupusnephritis.ArthritisRheum58: 1107–1115, 2008

17. Lau CM, Broughton C, Tabor AS, Akira S,Flavell RA, Mamula MJ, Christensen SR,Shlomchik MJ, Viglianti GA, Rifkin IR,Marshak-Rothstein A: RNA-associated auto-antigens activate B cells by combined B cellantigen receptor/Toll-like receptor 7 engage-ment. J Exp Med 202: 1171–1177, 2005

18. Lech M, Kulkarni OP, Pfeiffer S, Savarese E,Krug A, Garlanda C, Mantovani A, AndersHJ: Tir8/Sigirr prevents murine lupus bysuppressing the immunostimulatory effectsof lupus autoantigens. J Exp Med 205:1879–1888, 2008

19. Lech M, Skuginna V, Kulkarni OP, Gong J,Wei T, Stark RW, Garlanda C, Mantovani A,Anders HJ: Lack of SIGIRR/TIR8 aggravateshydrocarbon oil-induced lupus nephritis.J Pathol 220: 596–607, 2010

20. Leadbetter EA, Rifkin IR, Hohlbaum AM,Beaudette BC, Shlomchik MJ, Marshak-Rothstein A: Chromatin-IgG complexes acti-vate B cells by dual engagement of IgM andToll-like receptors.Nature416:603–607,2002

21. Guiducci C, Gong M, Xu Z, Gill M,Chaussabel D, Meeker T, Chan JH, WrightT,PunaroM,BollandS,SoumelisV,BanchereauJ, Coffman RL, Pascual V, Barrat FJ: TLR re-cognition of self nucleic acids hampers glu-cocorticoid activity in lupus. Nature 465:937–941, 2010

22. Patole PS, Zecher D, Pawar RD, Gröne HJ,Schlöndorff D, Anders HJ: G-rich DNAsuppresses systemic lupus. J Am SocNephrol 16: 3273–3280, 2005

23. Pawar RD, Ramanjaneyulu A, Kulkarni OP,Lech M, Segerer S, Anders HJ: Inhibition ofToll-like receptor-7 (TLR-7) or TLR-7 plusTLR-9 attenuates glomerulonephritis andlung injury in experimental lupus. J Am SocNephrol 18: 1721–1731, 2007

24. Teichmann LL, OlsML, KashgarianM, ReizisB, KaplanDH, ShlomchikMJ: Dendritic cellsin lupus are not required for activation of Tand B cells but promote their expansion,resulting in tissue damage. Immunity 33:967–978, 2010

25. Theofilopoulos AN, Baccala R, Beutler B,Kono DH: Type I interferons (alpha/beta) inimmunity and autoimmunity. Annu Rev Im-munol 23: 307–336, 2005

26. Chan OT, Hannum LG, Haberman AM,Madaio MP, Shlomchik MJ: A novel mousewith B cells but lacking serum antibody re-veals an antibody-independent role for Bcells in murine lupus. J ExpMed 189: 1639–1648, 1999

27. Zikherman J, Parameswaran R, Weiss A:Endogenous antigen tunes the responsivenessof naive B cells but not T cells. Nature 489:160–164, 2012

28. Allam R, Sayyed SG, Kulkarni O, LichtnekertJ, Anders HJ: Murine double minute-2drives systemic lupus erythematosus andlupus nephritis. J Am Soc Nephrol 22:2016–2027, 2011

29. Liu Z, Davidson A: BAFF and selection ofautoreactive B cells. Trends Immunol 32:388–394, 2011

30. Gayed M, Gordon C: Novel treatments forsystemic lupus erythematosus. Curr OpinInvestig Drugs 11: 1256–1264, 2010

31. Lech M, Weidenbusch M, Kulkarni OP, RyuM, Darisipudi MN, Susanti HE, Mittruecker

HW, Mak TW, Anders HJ: IRF4 deficiencyabrogates lupus nephritis despite enhanc-ing systemic cytokine production. J Am SocNephrol 22: 1443–1452, 2011

32. Caricchio R,McPhie L, Cohen PL: UltravioletB radiation-induced cell death: Critical roleof ultraviolet dose in inflammation and lu-pus autoantigen redistribution. J Immunol171: 5778–5786, 2003

33. Cornacchia E, Golbus J, Maybaum J,Strahler J, Hanash S, Richardson B: Hydral-azine and procainamide inhibit T cell DNAmethylation and induce autoreactivity.J Immunol 140: 2197–2200, 1988

34. RichardsonB, Scheinbart L, Strahler J, GrossL, Hanash S, Johnson M: Evidence forimpaired T cell DNA methylation in sys-temic lupus erythematosus and rheumatoidarthritis. Arthritis Rheum 33: 1665–1673,1990

35. Hughes GC: Progesterone and autoim-mune disease. Autoimmun Rev 11: A502–A514, 2012

36. Tojo T, Friou GJ: Lupus nephritis: Varyingcomplement-fixing properties of immuno-globulin G antibodies to antigens of cellnuclei. Science 161: 904–906, 1968

37. Jacob N, Stohl W: Autoantibody-dependentand autoantibody-independent roles for Bcells in systemic lupus erythematosus: Past,present, and future. Autoimmunity 43: 84–97, 2010

38. Yu F, Wu LH, Tan Y, Li LH, Wang CL, WangWK,Qu Z, ChenMH,Gao JJ, Li ZY, ZhengX,Ao J, Zhu SN, Wang SX, Zhao MH, Zou WZ,Liu G: Tubulointerstitial lesions of patientswith lupus nephritis classified by the 2003International Society of Nephrology andRenal Pathology Society system. Kidney Int77: 820–829, 2010

39. Weening JJ, D’Agati VD, Schwartz MM,Seshan SV, Alpers CE, Appel GB, Balow JE,Bruijn JA, Cook T, Ferrario F, Fogo AB,Ginzler EM, Hebert L, Hill G, Hill P, JennetteJC, Kong NC, Lesavre P, Lockshin M, LooiLM, Makino H, Moura LA, Nagata M; In-ternational Society of Nephrology WorkingGroupontheClassificationofLupusNephritisRenal Pathology Society Working Groupon the Classification of Lupus Nephritis: Theclassification of glomerulonephritis in syste-mic lupus erythematosus revisited.Kidney Int65: 521–530, 2004

40. Nowling TK, Gilkeson GS: Mechanisms oftissue injury in lupus nephritis. Arthritis ResTher 13: 250, 2011

41. Yung S, Chan TM: Autoantibodies and res-ident renal cells in the pathogenesis of lupusnephritis: Getting to know the unknown. ClinDev Immunol 2012: 139365, 2012

42. Mortensen ES, Rekvig OP: Nephritogenicpotential of anti-DNA antibodies againstnecrotic nucleosomes. J Am Soc Nephrol20: 696–704, 2009

43. HedbergA, Fismen S, Fenton KA, Fenton C,Osterud B, Mortensen ES, Rekvig OP:

J Am Soc Nephrol 24: 1357–1366, 2013 Pathogenesis of Lupus Nephritis 1363

www.jasn.org BRIEF REVIEW

Page 8: The Pathogenesis of Lupus Nephritis - Nephrology Now

Heparin exerts a dual effect onmurine lupusnephritis by enhancing enzymatic chroma-tin degradation and preventing chromatinbinding in glomerular membranes. ArthritisRheum 63: 1065–1075, 2011

44. Yung S, Cheung KF, Zhang Q, Chan TM:Anti-dsDNA antibodies bind to mesangialannexin II in lupus nephritis. J Am SocNephrol 21: 1912–1927, 2010

45. Zhao Z, Deocharan B, Scherer PE, OzeliusLJ, Putterman C: Differential binding ofcross-reactive anti-DNA antibodies to me-sangial cells: The role of alpha-actinin.J Immunol 176: 7704–7714, 2006

46. Mjelle JE, Rekvig OP, Van Der Vlag J,Fenton KA: Nephritogenic antibodies bindin glomeruli through interaction with ex-posed chromatin fragments and not withrenal cross-reactive antigens. Autoimmu-nity 44: 373–383, 2011

47. FentonKA, Tømmerås B,Marion TN, RekvigOP: Pure anti-dsDNAmAbs need chromatinstructures to promote glomerular me-sangial deposits in BALB/c mice. Autoim-munity 43: 179–188, 2010

48. Sekine H, Ruiz P, Gilkeson GS, Tomlinson S:The dual role of complement in the pro-gression of renal disease in NZB/W F(1)mice and alternative pathway inhibition.Mol Immunol 49: 317–323, 2011

49. Sekine H, Kinser TT, Qiao F, Martinez E,Paulling E, Ruiz P, Gilkeson GS, TomlinsonS: The benefit of targeted and selective in-hibition of the alternative complementpathway for modulating autoimmunity andrenal disease in MRL/lpr mice. ArthritisRheum 63: 1076–1085, 2011

50. Bao L, Haas M, Quigg RJ: Complementfactor H deficiency accelerates developmentof lupus nephritis. J Am Soc Nephrol 22:285–295, 2011

51. Niederer HA, Clatworthy MR, Willcocks LC,Smith KG: FcgammaRIIB, FcgammaRIIIB,and systemic lupus erythematosus. Ann N YAcad Sci 1183: 69–88, 2010

52. Allam R, Lichtnekert J, Moll A, Taubitz A,Vielhauer V, Anders HJ: Viral RNA and DNAtrigger common antiviral responses in me-sangial cells. J Am Soc Nephrol 20: 1986–1996, 2009

53. Fluer K, Allam R, Zecher D, Kulkarni O,Lichtnekert J, Schwarz M, Beutler B,Vielhauer V, Anders HJ: Viral RNA inducestype I interferon-dependent cytokine re-lease and cell death in mesangial cells viamelanoma-differentiation-associated gene-5: Implications for viral infection-associatedglomerulonephritis. Am J Pathol 175: 2014–2022, 2009

54. Hägele H, Allam R, Pawar RD, Anders HJ:Double-stranded RNA activates type I inter-feron secretion in glomerular endothelialcells via retinoic acid-inducible gene (RIG)-1.NephrolDialTransplant24: 3312–3318, 2009

55. Hägele H, Allam R, Pawar RD, Reichel CA,Krombach F, Anders HJ: Double-stranded

DNA activates glomerular endothelial cellsand enhances albumin permeability via atoll-like receptor-independent cytosolic DNArecognitionpathway.AmJPathol 175: 1896–1904, 2009

56. Fairhurst AM, Xie C, Fu Y, Wang A,Boudreaux C, Zhou XJ, Cibotti R, Coyle A,Connolly JE, Wakeland EK, Mohan C: TypeI interferons produced by resident renalcells may promote end-organ disease inautoantibody-mediated glomerulonephri-tis. J Immunol 183: 6831–6838, 2009

57. Anders HJ, Lichtnekert J, Allam R: Interferon-alpha and -beta in kidney inflammation.Kidney Int 77: 848–854, 2010

58. Triantafyllopoulou A, Franzke CW, SeshanSV, Perino G, Kalliolias GD, Ramanujam M,van Rooijen N, Davidson A, Ivashkiv LB:Proliferative lesions and metalloproteinaseactivity in murine lupus nephritis mediatedby type I interferons and macrophages.Proc Natl Acad Sci U S A 107: 3012–3017,2010

59. Anders HJ, Vielhauer V, Eis V, Linde Y,KretzlerM, Perez de LemaG, Strutz F, BauerS, RutzM,Wagner H,GröneHJ, SchlöndorffD: Activation of toll-like receptor-9 inducesprogression of renal disease in MRL-Fas(lpr)mice. FASEB J 18: 534–536, 2004

60. Rich SA: Human lupus inclusions and in-terferon. Science 213: 772–775, 1981

61. Allam R, Anders HJ: The role of innate im-munity in autoimmune tissue injury. CurrOpin Rheumatol 20: 538–544, 2008

62. Wang Y, Ito S, Chino Y, Goto D, MatsumotoI, Murata H, Tsutsumi A, Hayashi T, UchidaK, Usui J, Yamagata K, Sumida T: Lasermicrodissection-based analysis of cytokinebalance in the kidneys of patients with lupusnephritis.Clin Exp Immunol 159: 1–10, 2010

63. KulkarniO, AndersHJ: Chemokines in lupusnephritis. Front Biosci 13: 3312–3320, 2008

64. Kulkarni O, Pawar RD, Purschke W, EulbergD, Selve N, Buchner K, Ninichuk V, SegererS, Vielhauer V, Klussmann S, Anders HJ:Spiegelmer inhibition of CCL2/MCP-1 ame-liorates lupus nephritis in MRL-(Fas)lpr mice.J Am Soc Nephrol 18: 2350–2358, 2007

65. Pérez de Lema G, Maier H, Franz TJ,Escribese M, Chilla S, Segerer S, CamarasaN, Schmid H, Banas B, Kalaydjiev S, BuschDH, Pfeffer K, Mampaso F, Schlöndorff D,Luckow B: Chemokine receptor Ccr2 de-ficiency reduces renal disease and prolongssurvival in MRL/lpr lupus-prone mice. J AmSoc Nephrol 16: 3592–3601, 2005

66. Anders HJ, Belemezova E, Eis V, Segerer S,Vielhauer V, Perez de Lema G, Kretzler M,Cohen CD, Frink M, Horuk R, Hudkins KL,Alpers CE, Mampaso F, Schlöndorff D: Lateonset of treatment with a chemokine re-ceptor CCR1 antagonist prevents progres-sion of lupus nephritis in MRL-Fas(lpr) mice.J Am Soc Nephrol 15: 1504–1513, 2004

67. Chong BF, Mohan C: Targeting the CXCR4/CXCL12 axis in systemic lupus erythematosus.

Expert Opin Ther Targets 13: 1147–1153,2009

68. Bussolati B, Peri G, Salvidio G, Verzola D,Mantovani A, Camussi G: The long pen-traxin PTX3 is synthesized in IgA glomeru-lonephritis and activates mesangial cells. JImmunol 170: 1466–1472, 2003

69. Deban L, Russo RC, Sironi M, Moalli F,Scanziani M, Zambelli V, Cuccovillo I,Bastone A, Gobbi M, Valentino S, Doni A,Garlanda C, Danese S, Salvatori G, SassanoM,EvangelistaV,RossiB, ZenaroE,ConstantinG, Laudanna C, Bottazzi B, Mantovani A: Re-gulation of leukocyte recruitment by the longpentraxin PTX3. Nat Immunol 11: 328–334,2010

70. Lech M, Römmele C, Kulkarni OP, SusantiHE, Migliorini A, Garlanda C, Mantovani A,Anders HJ: Lack of the long pentraxin PTX3promotes autoimmune lung disease but notglomerulonephritis in murine systemic lu-pus erythematosus. PLoS ONE 6: e20118,2011

71. Chang A, Henderson SG, Brandt D, Liu N,Guttikonda R, Hsieh C, Kaverina N, UtsetTO, Meehan SM, Quigg RJ, Meffre E, ClarkMR: In situ B cell-mediated immune re-sponses and tubulointerstitial inflammationin human lupus nephritis. J Immunol 186:1849–1860, 2011

72. NeusserMA, LindenmeyerMT, Edenhofer I,Gaiser S, Kretzler M, Regele H, Segerer S,Cohen CD: Intrarenal production of B-cellsurvival factors in human lupus nephritis.Mod Pathol 24: 98–107, 2011

73. Espeli M, Bökers S, Giannico G, DickinsonHA, Bardsley V, Fogo AB, Smith KG: Localrenal autoantibody production in lupus ne-phritis. J Am Soc Nephrol 22: 296–305,2011

74. Bekar KW, Owen T, Dunn R, Ichikawa T,Wang W, Wang R, Barnard J, Brady S,Nevarez S, GoldmanBI, KehryM, Anolik JH:Prolonged effects of short-term anti-CD20B cell depletion therapy in murine systemiclupus erythematosus. Arthritis Rheum 62:2443–2457, 2010

75. Ramanujam M, Bethunaickan R, Huang W,Tao H, Madaio MP, Davidson A: Selectiveblockade of BAFF for the prevention andtreatment of systemic lupus erythematosusnephritis in NZM2410mice.Arthritis Rheum62: 1457–1468, 2010

76. Apostolidis SA, Crispín JC, Tsokos GC: IL-17-producing T cells in lupus nephritis. Lu-pus 20: 120–124, 2011

77. Bethunaickan R, Berthier CC, RamanujamM, Sahu R, Zhang W, Sun Y, Bottinger EP,Ivashkiv L, Kretzler M, Davidson A: A uniquehybrid renal mononuclear phagocyte acti-vation phenotype in murine systemic lupuserythematosus nephritis. J Immunol 186:4994–5003, 2011

78. Katsiari CG, Liossis SN, Sfikakis PP: Thepathophysiologic role of monocytes and mac-rophages in systemic lupus erythematosus:

1364 Journal of the American Society of Nephrology J Am Soc Nephrol 24: 1357–1366, 2013

BRIEF REVIEW www.jasn.org

Page 9: The Pathogenesis of Lupus Nephritis - Nephrology Now

A reappraisal. Semin Arthritis Rheum 39:491–503, 2010

79. Smeets B, Kuppe C, Sicking EM, Fuss A,Jirak P, van Kuppevelt TH, Endlich K,Wetzels JF, Gröne HJ, Floege J, MoellerMJ: Parietal epithelial cells participate in theformation of sclerotic lesions in focal seg-mental glomerulosclerosis. J Am SocNephrol 22: 1262–1274, 2011

80. Smeets B, Angelotti ML, Rizzo P, Dijkman H,Lazzeri E, Mooren F, Ballerini L, Parente E,SagrinatiC,MazzinghiB, RonconiE,BecherucciF, Benigni A, Steenbergen E, Lasagni L,Remuzzi G, Wetzels J, Romagnani P: Renalprogenitor cells contribute to hyperplasticlesions of podocytopathies and crescenticglomerulonephritis. J Am Soc Nephrol 20:2593–2603, 2009

81. Smeets B, Uhlig S, Fuss A, Mooren F,Wetzels JF, Floege J, Moeller MJ: Tracingthe origin of glomerular extracapillary le-sions from parietal epithelial cells. J Am SocNephrol 20: 2604–2615, 2009

82. Ryu M, Migliorini A, Miosge N, Gross O,Shankland S, Brinkkoetter PT, Hagmann H,Romagnani P, Liapis H, Anders HJ: Plasmaleakage through glomerular basementmembrane ruptures triggers the pro-liferation of parietal epithelial cells andcrescent formation in non-inflammatoryglomerular injury [published online aheadof print May 3, 2012]. J Pathol doi:10.1002/path.4046

83. Croca SC, Rodrigues T, Isenberg DA: As-sessment of a lupus nephritis cohort over a30-year period. Rheumatology (Oxford) 50:1424–1430, 2011

84. Houssiau F: Thirty years of cyclophospha-mide: Assessing the evidence. Lupus 16:212–216, 2007

85. Houssiau FA, Vasconcelos C, D’Cruz D,Sebastiani GD, de RamonGarrido E, DanieliMG, Abramovicz D, Blockmans D, Cauli A,Direskeneli H, Galeazzi M, Gül A, Levy Y,Petera P, Popovic R, Petrovic R, Sinico RA,Cattaneo R, Font J, Depresseux G, CosynsJP, Cervera R: The 10-year follow-up data ofthe Euro-Lupus Nephritis Trial comparinglow-dose and high-dose intravenous cyclo-phosphamide. Ann Rheum Dis 69: 61–64,2010

86. Kulkarni OP, Sayyed SG, Kantner C, Ryu M,Schnurr M, Sárdy M, Leban J, Jankowsky R,Ammendola A, Doblhofer R, Anders HJ:4SC-101, a novel small molecule dihy-droorotate dehydrogenase inhibitor, sup-presses systemic lupus erythematosus inMRL-(Fas)lpr mice. Am J Pathol 176: 2840–2847, 2010

87. Wang HY, Cui TG, Hou FF, Ni ZH, Chen XM,Lu FM, Xu FF, Yu XQ, Zhang FS, Zhao XZ,Zhao MH, Wang GB, Qian JQ, Cai GY, ZhuTY, Wang YH, Jiang ZP, Li YN, Mei CL, ZouWZ; China Leflunomide Lupus NephritisStudy Group: Induction treatment of pro-liferative lupus nephritis with leflunomide

combined with prednisone: A prospectivemulti-centre observational study. Lupus 17:638–644, 2008

88. Chan OT, Madaio MP, Shlomchik MJ: Thecentral and multiple roles of B cells in lupuspathogenesis. Immunol Rev 169: 107–121,1999

89. Gregersen JW, Jayne DR: B-cell depletionin the treatment of lupus nephritis. Nat RevNephrol 8: 505–514, 2012

90. Rovin BH, Furie R, Latinis K, Looney RJ,Fervenza FC, Sanchez-Guerrero J, MaciucaR, Zhang D, Garg JP, Brunetta P, Appel G;LUNAR Investigator Group: Efficacy andsafety of rituximab in patients with ac-tive proliferative lupus nephritis: The Lu-pus Nephritis Assessment with Rituximabstudy. Arthritis Rheum 64: 1215–1226,2012

91. Merrill JT, Neuwelt CM, Wallace DJ,Shanahan JC, Latinis KM, Oates JC, UtsetTO, Gordon C, Isenberg DA, Hsieh HJ,ZhangD, Brunetta PG: Efficacy and safety ofrituximab in moderately-to-severely activesystemic lupus erythematosus: The ran-domized, double-blind, phase II/III systemiclupus erythematosus evaluation of ritux-imab trial. Arthritis Rheum 62: 222–233,2010

92. Weidenbusch M, Römmele C, Schröttle A,Anders HJ: Beyond the LUNAR trial. Ef-ficacy of rituximab in refractory lupusnephritis. Nephrol Dial Transplant 28: 106–111, 2013

93. Navarra SV, Guzmán RM, Gallacher AE, HallS, Levy RA, Jimenez RE, Li EK, Thomas M,KimHY, LeónMG, Tanasescu C, Nasonov E,Lan JL, Pineda L, Zhong ZJ, Freimuth W,Petri MA; BLISS-52 Study Group: Efficacyand safety of belimumab in patients withactive systemic lupus erythematosus: Arandomised, placebo-controlled, phase 3trial. Lancet 377: 721–731, 2011

94. Manzi S, Sánchez-Guerrero J, Merrill JT,Furie R, Gladman D, Navarra SV, GinzlerEM, D’Cruz DP, Doria A, Cooper S, ZhongZJ, Hough D, Freimuth W, Petri MA; BLISS-52 and BLISS-76 Study Groups: Effects ofbelimumab, a B lymphocyte stimulator-specific inhibitor, on disease activity acrossmultiple organ domains in patients withsystemic lupus erythematosus: Combinedresults from two phase III trials. Ann RheumDis 71: 1833–1838, 2012

95. Mok CC: Abatacept for systemic lupuserythematosus: The outlook. Expert OpinBiol Ther 12: 1559–1561, 2012

96. Merrill JT, Burgos-Vargas R, Westhovens R,Chalmers A, D’Cruz D, Wallace DJ, Bae SC,Sigal L, Becker JC, Kelly S, Raghupathi K, LiT, Peng Y, Kinaszczuk M, Nash P: The effi-cacy and safety of abatacept in patientswith non-life-threatening manifestationsof systemic lupus erythematosus: Resultsof a twelve-month, multicenter, explor-atory, phase IIb, randomized, double-blind,

placebo-controlled trial. Arthritis Rheum 62:3077–3087, 2010

97. Hiepe F, Dörner T, Hauser AE, Hoyer BF,Mei H, Radbruch A: Long-lived autoreactiveplasma cells drive persistent autoimmuneinflammation. Nat Rev Rheumatol 7: 170–178, 2011

98. Ichikawa HT, Conley T, Muchamuel T, JiangJ, Lee S, Owen T, Barnard J, Nevarez S,Goldman BI, Kirk CJ, Looney RJ, Anolik JH:Beneficial effect of novel proteasome in-hibitors in murine lupus via dual inhibition oftype I interferon and autoantibody-secretingcells. Arthritis Rheum 64: 493–503, 2012

99. Neubert K, Meister S, Moser K, Weisel F,Maseda D, Amann K,Wiethe C,Winkler TH,Kalden JR, Manz RA, Voll RE: The protea-some inhibitor bortezomib depletes plasmacells and protects mice with lupus-like dis-ease from nephritis. Nat Med 14: 748–755,2008

100. Anders HJ: Pseudoviral immunity - a novelconcept for lupus. Trends Mol Med 15:553–561, 2009

101. Barrat FJ, Coffman RL: Development of TLRinhibitors for the treatment of autoimmunediseases. Immunol Rev 223: 271–283, 2008

102. Hahn BH, McMahon MA, Wilkinson A,Wallace WD, Daikh DI, Fitzgerald JD,Karpouzas GA, Merrill JT, Wallace DJ,Yazdany J, Ramsey-Goldman R, Singh K,Khalighi M, Choi SI, Gogia M, Kafaja S,KamgarM, LauC,MartinWJ, Parikh S, PengJ, Rastogi A, Chen W, Grossman JM; Amer-ican College of Rheumatology: AmericanCollege of Rheumatology guidelines forscreening, treatment, and management oflupus nephritis. Arthritis Care Res (Hoboken)64: 797–808, 2012

103. Clowse ME, Magder L, Witter F, Petri M:Hydroxychloroquine in lupus pregnancy.Arthritis Rheum 54: 3640–3647, 2006

104. Pons-Estel GJ, Alarcón GS, McGwin G Jr,Danila MI, Zhang J, Bastian HM, ReveilleJD, Vilá LM; Lumina Study Group: Pro-tective effect of hydroxychloroquine on re-nal damage in patients with lupus nephritis:LXV, data from a multiethnic US cohort.Arthritis Rheum 61: 830–839, 2009

105. Barrat FJ, Meeker T, Chan JH, Guiducci C,CoffmanRL: Treatment of lupus-pronemicewith a dual inhibitor of TLR7 and TLR9 leadsto reduction of autoantibody productionand amelioration of disease symptoms. EurJ Immunol 37: 3582–3586, 2007

106. Merrill JT, Wallace DJ, Petri M, Kirou KA,Yao Y, White WI, Robbie G, Levin R, BerneySM, Chindalore V, Olsen N, Richman L, LeC, Jallal B, White B; Lupus Interferon SkinActivity (LISA) Study Investigators: Safetyprofile and clinical activity of sifalimumab, afully human anti-interferon a monoclonalantibody, in systemic lupus erythematosus:A phase I, multicentre, double-blind ran-domised study. Ann Rheum Dis 70: 1905–1913, 2011

J Am Soc Nephrol 24: 1357–1366, 2013 Pathogenesis of Lupus Nephritis 1365

www.jasn.org BRIEF REVIEW

Page 10: The Pathogenesis of Lupus Nephritis - Nephrology Now

107. Aringer M, Burkhardt H, Burmester GR,Fischer-Betz R, Fleck M, Graninger W,Hiepe F, Jacobi AM, Kötter I, Lakomek HJ,Lorenz HM, Manger B, Schett G, SchmidtRE, Schneider M, Schulze-Koops H,Smolen JS, Specker C, Stoll T, StrangfeldA, Tony HP, Villiger PM, Voll R, Witte T,

Dörner T: Current state of evidence on‘off-label’ therapeutic options for sys-temic lupus erythematosus, includingbiological immunosuppressive agents,in Germany, Austria and Switzerland—a consensus report. Lupus 21: 386–401,2012

108. Kulkarni O, Eulberg D, Selve N, Zöllner S,Allam R, Pawar RD, Pfeiffer S, Segerer S,KlussmannS,AndersHJ:Anti-Ccl2Spiegelmerpermits 75% dose reduction of cyclophos-phamide to control diffuse proliferative lupusnephritis andpneumonitis inMRL-Fas(lpr)mice.J Pharmacol Exp Ther 328: 371–377, 2009

1366 Journal of the American Society of Nephrology J Am Soc Nephrol 24: 1357–1366, 2013

BRIEF REVIEW www.jasn.org