basic and translational concepts of immune-mediated glomerular

19
BRIEF REVIEW www.jasn.org Basic and Translational Concepts of Immune-Mediated Glomerular Diseases William G. Couser Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington ABSTRACT Genetically modied immune responses to infections and self-antigens initiate most forms of GN by generating pathogen- and danger-associated molecular patterns that stimulate Toll-like receptors and complement. These innate immune responses activate circulating monocytes and resident glomerular cells to release inamma- tory mediators and initiate adaptive, antigen-specic immune responses that collectively damage glomerular structures. CD4 T cells are needed for B celldriven antibody production that leads to immune complex formation in glomeruli, comple- ment activation, and injury induced by both circulating inammatory and resident glomerular effector cells. Th17 cells can also induce glomerular injury directly. In this review, information derived from studies in vitro, well characterized experimental models, and humans summarize and update likely pathogenic mechanisms involved in human diseases presenting as nephritis (postinfectious GN, IgA nephropathy, antiglomerular basement membrane and antineutrophil cytoplasmic antibodymediated crescentic GN, lupus nephritis, type I membranoproliferative GN), and nephrotic syndrome (minimal change/FSGS, membranous nephropathy, and C3 glomerulopathies). Advances in understanding the immunopathogenesis of each of these entities offer many opportunities for future therapeutic interventions. J Am Soc Nephrol 23: cccccc, 2012. doi: 10.1681/ASN.2011030304 Recent reviews of the immune mecha- nisms that lead to glomerular disease have been published elsewhere. 1,2 This review is organized by diseases rather than mechanisms to provide a transla- tional overview of how immune responses mediate the glomerular injury seen by clinicians and pathologists. The pro- cesses described derive from studies done in vitro and in an array of animal models of glomerular diseases as well as in humans. Cell cultures are not glomer- uli, and mice and rats are not humans, but experience has taught us that mech- anisms dened in these settings often translate into better understanding of similar processes seen in human disease. Schematic overviews of the major path- ogenic sequences currently believed to be operative in human GN and their in- teractions are presented in Figures 1 through 4. OVERVIEW OF BASIC IMMUNE MECHANISMS The Innate Immune Response Toll-Like Receptors Toll-like receptors (TLRs) are ancient and ubiquitous pattern recognition recep- tors present on all cell membranes and intracellularly between cytoplasm and en- dosomes (Figure 1). 36 TLRs recognize conserved immunostimulatory molecular patterns (antigens) like peptidoglycans, LPSs, and bacterial and viral nucleic acids (pathogen-associated molecular patterns [PAMPs]) as well as endogenous cell-derived patterns (danger-associated molecular patterns [DAMPs]). Another related cytoplasmic group of receptors called Nod-like receptors (NLRs) has re- cently been described. 6 TLR ligation is central to activating the non-antigen- specic innate immune system in im- mediate response to pathogens, but TLR activation is also required for adap- tive, antigen-specic immune responses by facilitating conversion of dendritic cells to antigen-presenting cells. 4 7 TLRs activate multiple signaling path- ways that lead to local release of a variety of cytokines, chemokines, and other in- ammatory mediators by all cells, includ- ing glomerular cells. 4,5 Thus, TLRs and NLRs connect initiating events with me- diation of tissue injury in GN associated with infections or autoimmunity or both. Complement The complement system and its regula- tory proteins are also ancient compo- nents of the innate immune system with multiple roles in human GN (Figure 2). 711 The innate immune response involves immediate complement activation through the mannose binding lectin (MBL) or alternative pathways. 7,10 Acti- vation of the MBL pathway proceeds Published online ahead of print. Publication date available at www.jasn.org. Correspondence: Dr. William G. Couser, 16050 169th Avenue NE, Woodinville, WA 98072. Email: [email protected] Copyright © 2012 by the American Society of Nephrology J Am Soc Nephrol 23: cccccc, 2012 ISSN : 1046-6673/2303-ccc 1

Upload: duongdung

Post on 02-Feb-2017

231 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Basic and Translational Concepts of Immune-Mediated Glomerular

BRIEF REVIEW www.jasn.org

Basic and Translational Concepts of Immune-MediatedGlomerular Diseases

William G. Couser

Division of Nephrology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington

ABSTRACTGenetically modified immune responses to infections and self-antigens initiatemostforms of GN by generating pathogen- and danger-associated molecular patternsthat stimulate Toll-like receptors and complement. These innate immune responsesactivate circulating monocytes and resident glomerular cells to release inflamma-tory mediators and initiate adaptive, antigen-specific immune responses thatcollectively damage glomerular structures. CD4 T cells are needed for B cell–drivenantibody production that leads to immune complex formation in glomeruli, comple-ment activation, and injury induced by both circulating inflammatory and residentglomerular effector cells. Th17 cells can also induce glomerular injury directly. In thisreview, information derived from studies in vitro, well characterized experimentalmodels, and humans summarize and update likely pathogenic mechanisms involvedin human diseases presenting as nephritis (postinfectious GN, IgA nephropathy,antiglomerular basement membrane and antineutrophil cytoplasmic antibody–mediated crescentic GN, lupus nephritis, type I membranoproliferative GN), andnephrotic syndrome (minimal change/FSGS, membranous nephropathy, and C3glomerulopathies). Advances in understanding the immunopathogenesis of eachof these entities offer many opportunities for future therapeutic interventions.

J Am Soc Nephrol 23: ccc–ccc, 2012. doi: 10.1681/ASN.2011030304

Recent reviews of the immune mecha-nisms that lead to glomerular diseasehave been published elsewhere.1,2 Thisreview is organized by diseases ratherthan mechanisms to provide a transla-tional overviewof how immune responsesmediate the glomerular injury seen byclinicians and pathologists. The pro-cesses described derive from studiesdone in vitro and in an array of animalmodels of glomerular diseases as well asin humans. Cell cultures are not glomer-uli, and mice and rats are not humans,but experience has taught us that mech-anisms defined in these settings oftentranslate into better understanding ofsimilar processes seen in human disease.Schematic overviews of the major path-ogenic sequences currently believed to

be operative in human GN and their in-teractions are presented in Figures 1through 4.

OVERVIEW OF BASIC IMMUNEMECHANISMS

The Innate Immune ResponseToll-Like ReceptorsToll-like receptors (TLRs) are ancientand ubiquitous pattern recognition recep-tors present on all cell membranes andintracellularly between cytoplasm and en-dosomes (Figure 1).3–6 TLRs recognizeconserved immunostimulatorymolecularpatterns (antigens) like peptidoglycans,LPSs, and bacterial and viral nucleicacids (pathogen-associated molecular

patterns [PAMPs]) as well as endogenouscell-derived patterns (danger-associatedmolecular patterns [DAMPs]). Anotherrelated cytoplasmic group of receptorscalled Nod-like receptors (NLRs) has re-cently been described.6 TLR ligation iscentral to activating the non-antigen-specific innate immune system in im-mediate response to pathogens, butTLR activation is also required for adap-tive, antigen-specific immune responsesby facilitating conversion of dendriticcells to antigen-presenting cells.4–7

TLRs activate multiple signaling path-ways that lead to local release of a varietyof cytokines, chemokines, and other in-flammatory mediators by all cells, includ-ing glomerular cells.4,5 Thus, TLRs andNLRs connect initiating events with me-diation of tissue injury in GN associatedwith infections or autoimmunity or both.

ComplementThe complement system and its regula-tory proteins are also ancient compo-nents of the innate immune system withmultiple roles inhumanGN(Figure2).7–11

The innate immune response involvesimmediate complement activationthrough the mannose binding lectin(MBL) or alternative pathways.7,10 Acti-vation of the MBL pathway proceeds

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

Correspondence: Dr. William G. Couser, 16050169th Avenue NE, Woodinville, WA 98072. Email:[email protected]

Copyright © 2012 by the American Society ofNephrology

J Am Soc Nephrol 23: ccc–ccc, 2012 ISSN : 1046-6673/2303-ccc 1

Page 2: Basic and Translational Concepts of Immune-Mediated Glomerular

whenMBL binds tomannose residues onpathogens and activates the serine pro-teases, MASP-1 and MASP-2, leading toactivation of C4 and C2. The alternativepathway is activated spontaneously byhydrolysis of C3 and amplified by defectsin complement regulation. Non-Ig zymo-gens such as damaged cells and bacterialand viral proteins can also activate thealternative pathway beginning directlyat C3. The same initiating event may ac-tivate more than one pathway.

Complement activation products arethe principal mediators of antibody-induced GN (Figures 1 and 2). Usuallythis involves C1q binding to Ig that leadsto classic pathway activation through C4and C2; however, some Igs, dependingon their level of glycosylation, can also

bind MBL. IgG subclasses 1 and 3 andIgM are classic complement pathway ac-tivators, whereas IgG 2 and 4, IgA, IgD,and IgE activate complement poorly.1,12

All complement activation pathwaysproceed through cleavage of C3 and C5leading to release of chemotactic factorssuch as C5a that attract inflammatorycells (neutrophils, macrophages, andplatelets) when abutting the circulationas well as formation of the terminalmembrane attack complex (C5b-9)(Figure 2).7–9 Sublytic quantities ofC5b-9 can insert into lipid bilayers ofadjacent glomerular cell membranes, ini-tiate several signaling pathways, and con-vert these cells to effector cells, whichmay proliferate; release a variety of cy-tokines, growth factors, eicosanoids,

oxidants, proteases, and other acute in-flammatorymediators; as well as upregu-late production of matrix componentsthat contribute to chronic scarring andsclerosis (Figure 1).1,12,13 Complementactivation products like C5a can also ac-tivate TLRs.14

Complement activation in vivo istightly regulated by a number of circu-lating and cell-bound complement regu-latory proteins (CRPs), whose functions,mutations and deficiencies are also im-portant in the development of severalglomerular diseases.8,9 Abnormalitiesin serum complement profiles are some-times helpful in assessing the nature ofthe underlying disease and its activity,but significant complement-mediatedinjury may occur locally without alter-ations in circulating complement com-ponents (Table 1).

The Adaptive Immune ResponseIgCD4 T helper cells stimulate B cells andplasma cells to make antibodies specificfor particular antigens (Figure 3). On thebasis of older studies of serum sicknessin rabbits induced by single (acute) orrepeated (chronic) injections of BSA,glomerular immune deposits have longbeen attributed to the passive trapping ofcirculating, soluble antigen-IgG anti-body complexes (ICs).15,16 Other studiesdone in antiglomerular antibody models(nephrotoxic nephritis; NTN) demon-strate that antibody deposition activatescomplement through the classic comple-ment pathway generating chemotacticfactors that attract circulating inflamma-tory effector cells, which then cause tissueinjury (Figures 1 and 3).17 Typical granu-lar IC deposits can also form locally, or insitu, due to antibody binding to either ex-ogenous planted antigens or endogenousglomerular components (Figure 3).18–23

There are several variables that determinethe biopsy findings and clinical conse-quences in IC GN: (1) where the depositsform—ICs of the same composition in asubendothelial distribution lead to exu-dative inflammatory cell infiltrates, inthe mesangium to mesangial cell pro-liferation and matrix expansion, andin a subepithelial distribution to a

Figure 1. Schematic overview of both the innate and adaptive immune mechanisms thatmediate tissue injury in GN. Etiologic events expose immunostimulatory PAMPs or DAMPsthat activate both the innate (red) and the adaptive (antigen specific, blue) immune sys-tems, which also interact with each other. Activation of the innate immune system occursimmediately and involves TLRs orNLRsonboth circulating inflammatory cells and residentglomerular cells. TLR activation results in release of inflammatory mediators that causeglomerular injury. Some PAMPs and DAMPs can activate complement directly through theinnate immune system. TLRs are also required to activate the adaptive immune systemthrough antigen-presenting cells that promote differentiation of CD4 helper cells, B cellactivation, and antibody production. Antibodies lead to circulating complex trapping orin situ formation of immune complexes that can activate both the TLR and complementcomponents of the innate immune system. Complement activation generates the che-motactic factor C5a that attracts circulating inflammatory cells (including neutrophils,macrophages, basophils, and natural killer cells), which release mediators and damageglomeruli and C5b-9 that activates resident glomerular cells to do the same. CD4 Th1 andTh2 cells cause tissue injury primarily thorough macrophages and basophils, respectively,whereas Th17 cells can mediate glomerular damage directly. CD4 regulatory cells (Tregs)downregulate the adaptive immune response.

2 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 3: Basic and Translational Concepts of Immune-Mediated Glomerular

noninflammatory lesion with podocyteinjury, effacement, and heavy protein-uria23–25; (2) the biologic properties of theantibody (or antigen) itself—particularlycomplement activating capacity, Fc re-ceptor affinity, ability to form lattices, orcryoprecipitability1,12,26; (3) the mecha-nism of deposit formation—when ICsform in situ, the process usually induceslocal tissue injury, whereas passive trap-ping of ICs formed in the circulation hasnot beenwell shown to be nephritogenic(Figure 3)1,22,23; and (4) the quantity—the more deposits form, the more severethe disease.

T CellsIn addition toprovidinghelp forBcells,27

some antigen-specific CD4 T cells alone,

sensitized to either self or nonself anti-gens that are localized in glomeruli, caninduce antibody-independent tissue in-jury.28,29 Although all subsets of T cellsare now implicated in GN, includingdendritic antigen-presenting cells(DCs) and CD4 helper cells of the Th1,Th2, and T regulatory (Treg) lineages,IL17-producing Th17 cells likely ac-count for much of T cell–induced in-flammation (Figure 4).30–34 Th17 cellsare attracted by mechanisms involvingchemokines and their receptors, and re-lease cytokines such as IL9, IL17, IL21,IL22, and TNFa, which induce othercells to produce additional proinflam-matory chemokines that attract neutro-phils and monocytes and also activateresident glomerular cells.30–33,35 Th17

cells are found in renal biopsies in severalforms of human GN.36 The T cell com-ponent of the adaptive immune responseis regulated by Tregs.27

Diseases Usually Presenting as GNPostinfectious or Poststreptococcal GNThe acute, diffuse exudative and pro-liferative lesion of poststreptococcal GN(PSGN) was long regarded as the humanequivalent of the acute, one-shot serumsickness model in rabbits leading to aprolonged search for the nephritogenicstreptococcal antigen. Although manycandidate proteins have been proposed,most have failed to meet strict criteriafor causality.37–39 However, streptococ-cal pyogenic exotoxin B (SpeB) meetsmost of these criteria,37,38 although ithas not been implicated in all cases ofepidemic PSGN.40 SpeB is a small (28kD), cationic (pK 9.3) cysteine prote-ase with complement-activating andplasmin-binding properties and repre-sents 90% of the secreted extracellularprotein made in vivo by nephritogenicstrains of group A streptococci.37,38 An-tibody to SpeB correlates with disease ac-tivity in PSGN and co-localizes with IgGand C3 in subepithelial humps.37–39,41

However, the intense exudative glomer-ular inflammatory response is not wellexplained by a serum sickness analogyand humps because circulating ICsdo not form subepithelial IC depositsdirectly and subepithelial IC depositsdo not produce inflammation.22–25

Moreover, IgG is sometimes absent oris only a minor constituent of the de-posits, whereas C3 deposition oftenboth precedes and exceeds detectableIgG.42,43

Possible explanations for these appar-ent contradictions include observationsthat some subendothelial deposits arealso present in PSGN,42,43 perhaps be-cause antibody to SpeB also exhibits mo-lecular mimicry with endothelial cells.44

In addition, SpeB alone can activatecomplement directly through the MBLpathway independent of IgG.37,38 SpeBalso exhibits plasmin-binding propertiesthat facilitate complement activation andmight cause proteolysis of glomerularbasement membrane (GBM), facilitating

Figure 2. Schematic depiction of the three pathways of complement activation as theyrelate to GN. In the adaptive immune system, complement-fixing antibodies (IgG1, IgG3,IgM) in immune complexes and CRP initiate classic pathway activation through C1q, C4,and C2 to form C4b2a, the classic pathway C3 convertase. In the innate immune system,PAMPs and DAMPs activate complement through the MBL or alternative pathways. Someinfectious PAMPs and DAMPs bind MBL leading to activation of MBL-associated serineproteases (MASPs), which activate C3 via C4, C2 and the classic pathway C3 convertase. Inthe alternative pathway, direct activation of C3 occurs spontaneously (“C3 tickover”) andby foreign surfaces, damaged cells, and IgA. Cleavage of C3 produces C3b, which com-bines with factor B and properdin to form the alternative pathway C3 convertase that isregulated by factors H and I to prevent excess C3 activation. Both classic and alternativepathway C3 convertases cleave C3 leading to release of C3b, which allows C5 convertaseformation. Cleavage of C5 produces the chemotactic factor C5a and C5b, which combineswith C6, C7, C8, and multiple C9 molecules to form the lipophilic C5b-9 membrane attackcomplex that can activate resident glomerular cells to become effector cells. C5b-9 for-mation is regulated by cell-bound CRPs such as CD59.

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 3

www.jasn.org BRIEF REVIEW

Page 4: Basic and Translational Concepts of Immune-Mediated Glomerular

the transit of dissociated subendothelialICs to form subepithelial humps.37,45,46

Finally, PSGN often exhibits autoim-mune features including both IgM andIgG rheumatoid factors with cryoglobu-lin activity, antiendothelial antibodies,anti-DNA antibodies, and antineutrophilcytoplasmic antibodies (ANCA), al-though their respective roles in mediat-ing the disease, if any, remain unclear(Table 1).47–50 Other forms of postinfec-tious GN such as those associated withendocarditis, infected ventricular-atrialshunts, visceral abscesses, and Staphylo-coccus aureus infectionwith IgA depositsare clearly mediated immunologically;however, the mechanisms involvedhave been explored in much lessdetail.39,41,51,52

IgA NephropathyIgA nephropathy (IgAN) is the mostcommon form of GN worldwide and ischaracterized by focal mesangial prolif-eration and matrix expansion accompa-nying diffuse mesangial deposits of IgA,and often IgG, C3, and C5b-9, usuallyassociatedwith recurrent episodes of GNthat often immediately follow mucosalviral infections.53–57 Although assumedto be mediated by mesangial trapping ofcirculating ICs, no exogenous antigenshave been identified consistently.55,56

IgA in mesangial deposits, and in ICform in the circulation, is polymeric

(mucosal) IgA1 that exhibits deficientO-linked glycosylation at five sites inthe hinge region of the molecule.55,56,58,59

The failure to normally glycosylate IgA1can be inherited in IgA nephropathy andHenoch-Schönlein purpura,60,61 but thedefect also seems to occur epigeneti-cally.62 Underglycosylated pIgA1 is pro-duced by mucosal B cells and might alsoreach the circulation if abnormal traffick-ing of these cells to the bone marrow oc-curs.63,64 Underglycosylated IgA1 predictsprogression and exhibits altered biologicproperties compared with normal IgA1including increased tendencies to self-aggregate, unmasking of MBL bindingsites leading to complement activation,binding to other molecules like fibronec-tin, IgG, and collagen IV. In circulatingmacromolecular form, it evades removalfrom the circulation by asialoglycopro-tein and CD 89 receptors, thus facilitat-ing mesangial localization.55,56,63,65–67 Itis not yet known if lanthanic mesangialIgA deposits seen in 6%–16% of normaldonor kidneys without disease containunderglycosylated IgA.

Although IgG autoantibodies to me-sangial cell antigens have been describedin IgAN,68 Suzuki et al. were the first toreport IgG antibodies directed to crypticantigenic structures in the hinge regionof the aberrantly glycosylated IgA1 mol-ecule (antiglycan antibodies), which cor-relate with disease activity.69 Antiglycan

antibodies form ICs with underglycosy-lated IgA1 that can be passively trappedin the mesangium.56 Alternatively, whenIgG antibody binds in situ to antigenicmaterial (or IgA1) in the mesangium,70

or on the mesangial cell membrane (theantithymocyte serum model in rats71–74),the mesangial cell response to acute im-mune injury closely simulates the clinicaland histopathologic features of humanIgAN.75–77

In IgAN, mesangial cells become ac-tivated through interactions between theIgA1 deposits and IgA Fca (CD89) re-ceptors, TLRs, and transferrin receptors(TfR, CD71).78,79 TLR activation by IgAaggregates, perhaps containing or ac-companied by PAMPs, may account forthe recurrent episodes of acute injurywith hematuria, particularly those thatimmediately follow infections.55,56,80

However, most experimental and clini-cal studies suggest a role for complementas well.7,9,81,82 C5b-9 generated fromcomplement activation induced by in-teraction of IgA1 aggregates with MBL,or in situ formation of ICs by IgG anti-glycan antibodies, inducesmesangial celltransformation to a-smooth muscleactin–expressing myofibroblast-likecells, upregulates genes for collagen typeI, and increases production of cyto-kines and growth factors such as IL1,IL6, TNFa, PDGF, TGFb, EGF, FGF,CTGF, andHGF, all resulting inmesangial

Table 1. Most common complement profiles and autoimmune features in GN

Disease Serum C Profile Autoimmune Features References

Poststreptococcal GN AP or MBL normalC1q, low C3-C9

Anti-C1q, IgG AECA*, anti-DNA, ANCA, proteindisulfide Isomerase (PDI), cardiac myosin

38,44,47–50

IgAN Normal Antiglycan, endothelial cell, mesangial cell, IgG, C1q 54,56,59Anti-GBM nephritis Normal Anti-GBM, ANCA (20%), anti-C1q 87,90,94,96ANCA-positive GN Normal Anti-MPO, PR3, cPR3, NET, DNA, endothelial cell,

? LAMP2119–122,124,133,134

Lupus nephritis CP, low C1q-C9 Anti-dsDNA, annexin, MPO, PR3, nucleosome, IgG,C1q, C1s,C1-INH, C4, cardiolipin, MBL, NET,H-ficolin, C3Nef

17,171–173

MPGN I CP, low C1q-C9 Anti-C3 Nef, C4 Nef, C1q 199,282MCD/FSGS Normal None 216,246,217,215Membranous nephropathy Normal Anti-PLA2R, DNA, NEP, aldose reductase, SOD2, C1q 83,268,255,257,260,281DDD AP, normal C1q,

low C3-C9Anti-C3Nef, C4 Nef, CFH, factor B, C1q 7,198,283,284,290

C3 nephropathy AP, normal C1q,low C3-C9

C3Nef, CFH, factor B 7,286

CP, classic pathway; AP, alternastive pathway; MBL, mannose binding lectin pathway; LAMP2, lysosomal membrane protein 2.

4 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 5: Basic and Translational Concepts of Immune-Mediated Glomerular

cell proliferation and matrix expan-sion.13,14,72–74,76–79,83 The pattern ofglomerular complement deposition inIgAN includes MBL, C4d, and C5b-9(but not C1q) that co-localize with IgA1and suggests both MBL and AP ratherthan classic pathway activation.81,82,84–86

Complement deposits correlate withboth disease severity and prognosis.84–86

Rapidly Progressive, Crescentic GNAnti-GBM NephritisAnti-GBMnephritis is characterized ini-tially by an acute, focal necrotizing GNwith crescents and linear deposition ofIgG,usuallywithC3, along theGBM.87,88

When associated with pulmonary alveo-lar hemorrhage, it is called Goodpas-ture’s syndrome. The role of anti-GBMantibody deposition inducing comple-ment activation, chemotactic factor re-lease, and neutrophil-mediated injurywas defined in NTN models in the1960s,89 and the pathogenicity of humananti-GBM antibody was confirmed bythe classic primate transfer studies ofLerner et al. in 1967.90 Studies in C32/2

and C42/2 mice implicate primarily theclassical complement pathway90–92 acti-vated by IgG1 and IgG3 anti-GBM anti-body that correlates with disease activityand recurrence in transplants.87,93,94 An-tibodies with apparently similar reactiv-ity (but with lower titers, lower avidity,and primarily of the IgG2 and IgG4subclasses) can be present in healthyhumans.95

GBM antigens are also expressed inseveral extrarenal tissues where they aresequestered by an endothelial cell layerimpermeable to IgG.96 The unique fe-nestrated endothelium in glomeruli al-lows free access of IgG to GBM. TheGBM antigen itself consists of two nor-mally sequestered or cryptic epitopes,EA and EB, residing on the noncollage-nous domain of both the a3 and a5chains of the NC1 hexamer of type IVcollagen.96,97 Antibody deposition re-quires perturbation of the quaternarystructure of the a3, a4, a5NCI hexamer,possibly initiated by oxidant injury,which results in a conformational changein the a3NCI and a5NCI domains (anautoimmune conformeropathy).96,98 In

rodent models, the nephritogenic GBMantigen has been mapped to as few asthree amino acid sequences in a core res-idue,99 but both intermolecular and in-tramolecular epitope spreading occur,suggesting that immune reactivity mayextend beyond the initial inducing auto-antigen.100 Pulmonary toxins such asinfections, smoke, and volatile hydro-carbons may damage endothelium andexpose antigen in alveolar capillariesaccounting for the pulmonary manifes-tations in Goodpasture’s syndrome.87,96

Whether such extrarenal events haveany role in autoimmunization is notknown.

T cell reactivity to GBM antigens wasfirst demonstrated 40 years ago, and apathogenic role for GBM antigen-specificsensitized T cells was proposed101 butgiven little credence at the time.102 How-ever, many subsequent studies haveconfirmed these original observationswith newer technologies103 and docu-mented that nephritogenic GBM anti-gens can induce a T cell–mediated GNwith crescents, proteinuria, and decreasedrenal function in the absence of anti-GBMantibody.29,104–107 The IL23/Th17 axis iscentral to the mediation of injury in anti-GBM models.31–34,36,108 Another uniquefeature of the T cell response to GBM isthe appearance of long-lived Tregs andinversion of the T cell effector/regulatorycell ratio later in the disease that may ac-count for why recurrences of anti-GBMdisease are uncommon compared withother autoimmune glomerulonephritidesin which Treg activity is often im-paired.109,110

The anti-GBM immune response inhumans is strongly linked to HLA DRB1alleles 1501, 0701, and 0101 with 1501conferring a relative risk ratio .8,whereas 0701 and 0101 are protec-tive.109,110 Possible triggering eventsinclude preceding infections or environ-mental toxins that might expose antigenicdeterminants in extrarenal tissue. Mostpatients have anti-GBM antibodies inthe circulation that predate clinical dis-ease.111 The disease can also be inducedexperimentallywith a small nephritogenicT cell epitope, pCol28-40, from thea3NC1 domain, which exhibits molecular

mimicry with PAMPs in some Gram-negative bacteria, especially Clostridiabotulinum.112 Finally, recent studies in-dicate that glomerular-derived antigenicpeptides that enter the urine can betaken up and degraded by tubular cellsand then presented to interstitial den-dritic cells leading to induction of animmune response in regional lymphnodes.113–116 The occurrence of ANCAantibodies and signs of vasculitis inup to 20% of anti-GBM patients, andexamples of anti-GBM disease occur-ring with membranous nephropathy,suggest that some of the proposed etio-logic factors in these diseases are op-erative in anti-GBM disease as well(Table 1).111,117,118

ANCA-Associated GNNecrotizing crescentic GN without im-mune deposits, later called pauci-immuneGN, was described in 1979,119 and adecade later linked to ANCA directedagainst myeloperoxidase (MPO) andproteinase 3 (PR3).120 It is character-ized by a focal necrotizing and crescen-tic GN with large gaps in the capillarywall associated with a smoldering, ne-phritic clinical course, usually in olderindividuals whomay also exhibit extra-renal vasculitic disease.120–122 The ma-jor entities associated with ANCA andGN are granulomatosis with polyangiitis(formerly known as Wegener’s granu-lomatosis),123 Churg-Strauss syn-drome, and microscopic polyangiitis,which may be renal-limited.124 Explo-rations of how anti-MPO and PR3antibodies mediate GN without depos-iting in glomeruli have defined entirelynew paradigms of immune glomerularinjury.125–128

In vitro studies show that cytokines,released in response to infections, primeneutrophils and upregulate adhesionmolecules on neutrophils and endothe-lial cells (L and E selectins, respectively)to facilitate localization in glomerularcapillaries.129,130 Cytokine-primed neu-trophils redistribute cytoplasmic pri-mary granules containing MPO andPR3 to the cell surface where ANCAIgG binds directly or through Fc, Fab’2,or neutrophil-specific Mac-1 receptors

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 5

www.jasn.org BRIEF REVIEW

Page 6: Basic and Translational Concepts of Immune-Mediated Glomerular

activating a respiratory burst with re-lease of cationic MPO and PR3 as wellas other proteases and oxidants.129–135

Neutrophil extracellular traps (NETs)are also formed containing entrappedMPO, PR3, andMPODNA in a chroma-tin web and these can mediate injury di-rectly through TLRs as well as modulatethe immune response.134,136 In ANCA-GN, NETs are present in the circulationand in glomeruli co-localized with neu-trophils and DCs, and anti-NET anti-bodies are present along with circulatingMPO-DNA complexes (nucleosomes).134

Activation of TLR2 and TLR9 exacer-bate experimental crescentic GN.136

MPO can also cause glomerular injurydirectly through oxidative mechanismsinvolving the MPO-H2O2-halide sys-tem resulting in halogenation of glo-merular structures and severe glomerularinjury.137

In 2002, Xaio et al. provided the firstcompelling in vivo evidence for ANCApathogenicity by transferring spleencells from an MPO null mouse immu-nized with murine MPO to an immuno-logically compromised host to induce aT cell–independent crescentic GN withproteinuria and reduced renal func-tion.138 Similar studies implicate animmune response to PR3 in pathogene-sis.139 Other models have utilizedtransfer of MPO+ bone marrow,140 ad-juvants that enhance the immune re-sponse and increase cytokine levels,141

and mice with subclinical GN immu-nized to humanMPO inwhich the cres-centic GN that follows is mediated bythe immune response to endogenousMPO.142 Studies of the Xaio modelconfirm neutrophil dependence and,despite the absence of antibody deposits,a requirement for alternative comple-ment pathway activation involving C5aand C5a receptors.7–10,143–147 Both al-ternative complement pathway proteinsand C5b-9 deposits are found in glomer-uli in human disease.147

Two other ANCA antigens have alsobeen studied. Lysosomalmembrane pro-tein 2 exhibits molecular mimicry withthe Fim H group of adhesins on someGram-negative bacteria and is expressedon endothelial cells and neutrophils.

Figure 3. Mechanisms of glomerular immune deposit formation. (A) Glomerular immunedeposit formation secondary topassive trappingof circulating immunecomplexes.Antigen(blue dots) antibody (green) complexes are forming in slight antigen excess. Soluble im-mune complexes formed in the circulation are thenpassively trapped in subendothelial andmesangial areasof theglomerulus,where they form lattices andenlarge tobecome immunedeposits detectable by immunofluorescence and electron microscopy. (B) Glomerularimmune deposit formation secondary to in situ formation of immune deposits. In the firstphase, cationic antigens (blue) localize independently of antibody in subendothelial ormesangial sites (larger antigens) or beneath podocytes in the subepithelial space (smallerantigens). In the second phase, free antibody binds to these planted antigens to formimmune complexes in situ. (C) Glomerular in situ immune deposit formation due toautoantibodies to normal glomerular constituents (triangles). Antigens depicted areGoodpasture’s GBM antigen (red), mesangial antigens such as annexin (green), endo-thelial antigens such as human lysosomal membrane protein 2 (brown), and podocyteantigens such as PLA2R and NEP (blue).

6 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 7: Basic and Translational Concepts of Immune-Mediated Glomerular

Lysosomal membrane protein 2 anti-bodies correlate with disease activityand induce a focal necrotizing and cres-centic GN without immune deposits inanimals.148 However, these intriguingobservations require further confirma-tion. An antibody directed against a 13amino acid sequence in complementaryPR3 (cPR3),149,150 encoded by the anti-sense strand of PR3 DNA, has been de-tected in a minority (20%) of ANCApatients.151,152 Anti-cPR3 IgG elicits ananti-idiotypic antibody response thatis reactive with native (sense) PR3, sug-gesting a role for autoantigen com-plementarity in initiating the disease.Because amino acid sequences in cPR3also have homologies with several bac-teria and viruses, this could representanother link to potentially etiologic in-fectious agents and the innate immunesystem.151 Anti-cPR3 antibodies are alsoreactive with plasminogen and delaydissolution of clots in vitro, potentially

contributing to the prominent fibrin de-position seen in ANCA GN.121

Other groups reason that the absenceof antibody deposits in ANCA-positiveGN, the limited correlation betweenANCA levels and disease activity, andthe absence of any detectable ANCA inapproximately 10%–20% of patientswith typical microscopic polyangiitis153

suggest a primary role for antibody-independent, T cell–mediated immunemechanisms.106,154–156 Consistent withthis hypothesis are persistent activationof T cells and elevation of soluble T cellproducts that correlate with disease ac-tivity,127,157,158 the prominence of tradi-tional Th1 delayed-type hypersensitivitymarkers like T cells, macrophages, fi-brin, and occasional granulomas inANCA-positive GN156 and T cell reactiv-ity to ANCA antigens in some pa-tients.159–162 T cells alone, includingTh17 cells, induce focal necrotizing andcrescentic GN when sensitized to a

planted glomerular antigen as might oc-cur with planted cationic MPO.28,161 Arecent study used combinations of miceselectively deficient in T cells, B cells, orMPO to demonstrate that active immu-nizationwith humanMPO (inmice withsubclinical glomerular injury) inducescrescentic GN without immune depositsthat requires the presence of endogenousMPO and T cell reactivity to MPO, butdoes not require B cells or anti-MPO an-tibody.142 Th17 cells and IL17a, as wellas TLRs 2 and 9, are also essential to thedevelopment of GN in a T cell–dependentmodel.136,161

Proposed etiologic agents in ANCAdisease include environmental toxinssuch as silica and infectious agents,including Gram-positive (S. aureus)and Gram-negative (Fim H adhesins)bacteria, viral infections, and severaldrugs.122,124–128 There also have been sig-nificant but low-level associations withpotential susceptibility genes and theirpolymorphisms, including ANCA anti-gens, HLA, immune response proteins,Fc receptors, cytokines and others, butno high-level associations have been de-scribed,163 other than DRB1*15 in AfricanAmericans.164 The relatively frequent ob-servation of ANCA antibodies in otherautoimmune glomerular diseases in-cluding anti-GBM disease, lupus, andmembranous nephropathy suggeststhat common etiologic or susceptibilityfactors may be present (Table 1).165–167

Lupus NephritisIn lupus nephritis, IgG, IgM, IgA (fullhouse), and C3 deposits are localizedprimarily in the mesangium in milddisease (mesangial lupus nephritis, classI and II), along the subendothelial aspectof the capillary wall with increasing pro-liferative/inflammatory lesions (focal ordiffuse proliferative lupus nephritis,class III and IV), or in the subepithelialspace with membranous lupus nephritis(class V).168,169 The autoimmune respon-ses that underlie lupus have been exten-sively studied in humans and in mousestrains that spontaneously develop thedisease and are beyond the scope ofthis review.170–173 The best-established

Figure 4. The T cell component of the adaptive immune system in GN. Antigen is pre-sented to naïve CD4 T cells by dendritic cells (signal 1). Depending on the predominatecytokine environment, T cells differentiate into CD4 T cell subsets that play different rolesin the pathogenesis of glomerular disease. In the presence of ΤGFb, Tregs develop thatmake TGFb, IL10, and CTLA4 that downregulate and control the immune response. IL12stimulates differentiation into Th1 cells that make IFNg and TNF and produce traditionalT cell/macrophage–mediated delayed-type hypersensitivity reactions. IL2, IL4, and IL13favor development of Th2 cells that make IL4, IL5, and IL13 and lead to allergic-type hy-persensitivity reactions involving IgE and eosinophils. The CD4 T cells most implicated inthe pathogenesis of GN are Th17 cells that differentiate in the presence of TGFb, IL6, andespecially IL17 and produce IL17a and IL21 that facilitate recruitment of other in-flammatory cells and can also cause tissue injury directly.

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 7

www.jasn.org BRIEF REVIEW

Page 8: Basic and Translational Concepts of Immune-Mediated Glomerular

functional immune abnormalities inlupus are loss of tolerance to numerousself-antigens, B cell hyperactivity withoverproduction of autoantibodies, anddefective T cell regulation.170

The most prominent serologic fea-ture of lupus is the presence of IgGanti-double-stranded DNA antibodies(anti-DNA) in serum and in glomeru-lar deposits.170,174 The deposits are usu-ally attributed to DNA–anti-DNA ICstrapped from the circulation, althoughinfusing anti-DNA or DNA–anti-DNAICs has not achieved either significantglomerular capillary wall localization ordisease expression in vivo.22,170,175

Somemonoclonal anti-DNA antibodiesexhibit cross-reactivity with capillarywall antigens, especially laminin anda-actinin176,177 and may become inter-nalized by cells within caveola, achievenuclear localization, and directly altercell functions including apoptosis.178

Mesangial deposits have also been asso-ciated with antibody to mesangial cellannexin, which co-localizes with IgGand C3 and correlates with disease ac-tivity.179 However, most recent studiesconclude that deposited anti-DNA re-acts with extracellular DNA in theform of nucleosomes that consist ofan anionic segment of DNA woundaround a highly cationic histone core,giving the structure a net positive chargeand thereby a high affinity for glomerularanionic sites.171,172 Defective apoptosisin SLE, perhaps related to an acquireddefect in DNAse I, leads to necrosis andrelease of chromatin debris from apopto-tic blebs allowing access of nucleosomesto antigen-presenting DCs as well as en-try into the circulation.170–172,180 Cir-culating nucleosomes are abundant inpatients with lupus nephritis, antinucleo-some antibodies correlate with disease,and both are present in membrane-associated electron-dense deposits.170–172

Although this could represent an epiphe-nomenon, nucleosomes are required foranti-DNA antibody localization to occurin glomeruli.171,172 Whether they localizeinitially as free antigenic material to initi-ate in situ IC formation or are trapped aspreformed ICs is not known. Nucleo-somes exhibit several other relevant

biologic properties, including the abilityto activate dendritic cells through bind-ing to TLRs 2 and 9, and they likely di-rectly activate resident glomerular cellsthrough TLRs as well.181,182 In that ca-pacity, they may mimic infectious non-self structures to generate DAMPs thatcould lead to both loss of tolerance andlocal inflammation.182

Other non-nucleosome autoanti-bodies have also been implicated in dif-ferent aspects of the renal lesions in lupus,particularly lupus anticoagulant, anticar-diolipin, antiphospholipid, and anti-b2glycoprotein I antibodies in glomerularmicrothrombosis, as well as anti-C1qantibodies, mixed cryoglobulins con-taining rheumatoid factors, and others(Table 1).170,183,184 Recent studies inboth experimental and human lupus alsoimplicate the Th2 immune response withB cell differentiation, activation of baso-phils, and production of IgE anti-DNAan-tibodies that deposit in glomeruli.185 B cellactivating factor (BAFF or BLyS), a cyto-kine of the TNF ligand superfamily thatactivates B cells and modulates the im-mune response by inhibiting B cell ap-optosis, is increased in lupus, likelycontributes to autoantibody produc-tion, and has recently become a poten-tial therapeutic target.186

The subepithelial immune deposits inclassV (membranous) lupus nephritis169

could result from dissociation of suben-dothelial ICs with transit across GBM toreform in a subepithelial location45,46 orfrom deposition of other lupus autoanti-bodies with specificity for podocyteantigens as occurs in idiopathic mem-branous nephropathy (see below).

Complement activated by IC depositsis a major mediator of tissue injury inlupus nephritis through both intracap-illary generation of neutrophil and mac-rophage chemotactic factors (class II–IV)and formation of C5b-9 (class V).7,9,10,187

Disease severity is reduced in murinemodels that lack selected complementproteins and is increased with deficientregulatory proteins.184,187,188 Blockingstudies in murine models suggest thatthe AP of complement is more importantin mediating kidney damage than theclassic pathway.189 The observation

that deficiencies of classic pathway pro-teins C1.C4.C2 are associated withincreased risk for lupus suggests protec-tive roles for complement as well.9,10,187

For example, 90% of patients with in-herited Ciq deficiency develop lupus, andC1q is produced by dendritic cells and in-volved in tolerance induction and clear-ance of both apoptotic cells and ICs.9,10

T cells exhibit complex and abnormalphenotypes in lupus.170,171,173 ActivatedT cells are expanded, provide excess helpto B cells, localize in renal cell infiltrates,and produce IL17, which correlateswith disease activity, all implying CD4and Th17 cell involvement.170,171,190

Antigen-specific T cell reactivity to nu-clear antigens is well documented inlupus nephritis,190 and Th17 cells andIL17 are increased in human and mu-rine SLE and correlate with diseaseactivity.170,171,173,191 IL17-producing Tcells, either Th17 or CD42CD82 (doublenegative) T cells, are present in nephritickidneys, and decreasing IL17 productionimproves murine lupus nephritis.170,191

In addition to increased CD4 activityin SLE, most studies also suggest an ac-companying defect in T regulatory cellactivity.170,171,173,192

The epigenetic events that induceautoimmunity in lupus include environ-mental exposures such as ultraviolet lightand certain drugs and viral infections,especially Epstein–Barr virus.170–173

Some of these interact with the immunesystem through inhibition of DNAmethylation, which can lead to over-expression of some genes resulting inhypomethylated CD4 cells, overpro-duction of some cytokines and Mdm2,and overproduction of IgG by B cells.193

There also are sufficiently well estab-lished co-occurrences of lupus nephritiswith other GNs, includingANCA-positiveGN,166 IgA,194 membranous nephropa-thy,169 and even a minimal change-likepodocytopathy195 to suggest commonetiologic factors (Table 1).

Type I Membranoproliferative GNType I membranoproliferative GN(MPGN I) has many clinical and patho-logic similarities to a renal-limited lupus

8 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 9: Basic and Translational Concepts of Immune-Mediated Glomerular

nephritis, including frequent autoanti-bodies such as rheumatoid factors andantinuclear, anticardiolipin, anti-C1q,anti-C3 convertase (C3Nef), and anti-endothelial antibodies (Table 1).196–199Hy-pocomplementemiawith a classic pathwayprofile, and increased disease susceptibilityin the presence of C2 and C4 deficiency, isalso common to both entities.7,9,10,188,200

The histologic features of capillary wallthickening, cellular proliferation, and infil-trating inflammatory cells associated withprimarily mesangial and subendothelialdeposits of IgG, IgM, and C3 are similarto lupus nephritis and are also seen in avariety of chronic neoplasias (especiallymonoclonal gammopathies), infections,and other autoimmune processes.196–199,201

However, in contrast to lupus, MPGN Iin adults is seen almost exclusively(.90%) in association with hepatitis Cviral (HCV) infection, and the glomerulardeposits often have prominent ultrastruc-tural features of cryoglobulins.202–204

The principal nephritogenic HCVantigen seems to be non-envelopedHCV E2 core protein, which is demon-strable in circulating ICs and in glomer-ular deposits.205–207 IgG3 antibodybound to HCV E2 can interact with theglobular domain of C1q, engage B cellsthrough both B cell receptors and TLR7,and elicit production of monoclonalIgMk antibody to polyclonal anti-HCVIgG (rheumatoid factor).196–199,208 Thesesoluble, but cryoprecipitable, aggregatesof IgG, IgM, viral proteins/nucleic acids,and C1q constitute the mesangial andsubendothelial immune deposits foundin glomeruli and cause local inflammationthrough direct interaction with TLRs 3, 7,and 9 on both infiltrating inflammatorycells and/or resident glomerular cells aswell as by inducing more classic pathwayC activation.196–198,209–213 As in lupus, thesubepithelial deposits often seen inMPGNI (and sometimes referred to as type IIIMPGN) may represent subendothelialdeposits that dissociate and reform insitu or autoantibodies to as yet uniden-tified podocyte antigens.45,46

As in lupus, complement likely playsboth nephritogenic and protective rolesinMPGNI.C1q seems tobe important inmediating the initial interaction between

IgM, IgG, HCV complexes, B cells, andTLRs,196,197,212 and complement activa-tion by immune deposits through theclassic pathway likely aggravates tissueinjury,7,10 although overexpression of acomplement regulatory protein, Crry,in a well studied murine model did notsignificantly ameliorate the disease.214

The roles of CD4 effector cells and Tregsin MPGN I are not yet well defined ineither animal models or in humans.

Diseases That Usually Present withNephrotic SyndromeMinimal Change Disease/IdiopathicFSGS SpectrumThere aremany clinical and pathogeneticobservations in minimal change disease(MCD)andidiopathicFSGS,whichsuggestthat they may represent different points onthe same disease spectrum. Some patientswithMCDare steroid resistant anddevelopFSGS, whereas some patients with biopsy-documented FSGS are steroid responsiveand behave like MCD.215–217 Both can betriggered by multiple initiating events, in-cluding infections, drugs, malignancies,and others.215–217 Both are diseases of thepodocyte that have been associated withcirculating permeability factors,215,217–220

can recur immediately in transplants,221,222

and can resolve when affected kidneysare placed in normal environments.223

Thus, differences in disease phenotypeand clinical expression could reflect varia-tion in the quantity of a commonmediatoror group of mediators.

Alternatively, mutations or epigeneticdifferences in podocyte genes that alterresponse to, or recovery from, such cir-culating mediators might also accountfor differences between MCD and FSGS.Mutations in podocyte genes that regu-late the slit diaphragm, cell membrane,and cytoskeleton are increasingly recog-nized, not only in FSGS but in otherformsofGNaswell.224AfricanAmericanswith nondiabetic nephropathy express var-iants in the gene encoding APOL1.225–229

Both clinical and experimental studiesdocument the importance of severalother genes, especially ones that regu-late the podocyte actin cytoskeleton,in modulating the development of pro-teinuria, foot process effacement, and

sclerosis, including RhoA, urokinasereceptor, Pdlim2, and connective tissuegrowth factor.230–234 Experimentally,podocyte expression of angiopoietin-like-4 is upregulated in experimentalMCD and responds to steroids.235 Al-ternatively, the possibility that MCDand FGS could involve entirely differ-ent pathogenetic mechanisms actingon normal podocytes has not beenexcluded.

Some evidence suggests that bothMCD and idiopathic FSGS reflect theeffect on podocytes of circulating, per-haps T cell–derived, non-IgG perme-ability factors,215,217–220 as suggestedfirst by Shalhoub in 1974.218 Studies byMcCarthy et al. demonstrate a factor inthe serum of patients with recurrentFSGS that alters the albumin reflec-tion coefficient of normal glomeruli invitro.220 In MCD, Koyama et al. showedthat factors secreted by T cell hydridomasderived from patients with active MCDtransfer a MCD-like lesion to normalrats.219 Despite these in vitro and in vivoobservations, identification of the re-sponsible factor(s) has proven frustrat-ingly elusive.220 Many cytokines andother mediators—including hemopexin,soluble podocyte urokinase receptor,TNFa, IL13, angiopoietin-like 4, andcardiotrophin-like cytokine 1—are in-creased in patients with MCD or FSGSand several also increase glomerular al-bumin permeability in vitro.231,236–238

Soluble urokinase receptor has been im-plicated in activating podocyte b3 integ-rins leading to FSGS,231 and mountingevidence suggests that increased plasmalevels of soluble podocyte urokinase re-ceptor mediate proteinuria in both activeand recurrent FSGS (but not MCD)through a similar integrin-related mecha-nism.230 Neutralization of cardiotrophin-like cytokine 1 reduces permeability factoractivity in FSGS serum as does galactoseand normal serum and urine.215–217

Human studies document Th2 polar-ization and elevated levels of IL13, a Th2cytokine with podocyte receptors, inactive MCD.236,237 IL13 alters podocytefunction238,239 and overexpression ofIL13 induces albuminuria and foot pro-cess effacement.240 Transfer of CD34+

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 9

www.jasn.org BRIEF REVIEW

Page 10: Basic and Translational Concepts of Immune-Mediated Glomerular

stem cells from patients with activeMCD also transfers proteinuria andcauses podocyte foot process efface-ment, although the responsible factoris unclear.241 CD80 (B7.1) is a T cellco-stimulatory molecule involved in an-tigen processing that is also expressed onpodocytes. Podocyte CD80 activationthrough TLR 3 and 4, independent ofT cells, causes proteinuria and foot pro-cess effacement.242,243 Recent studies byGarin et al. document increased levels ofCD80 in podocytes and in urine in activeMCD, but not FSGS,244 although mea-surement of urinary mRNA encodingCD80 demonstrates higher levels inFSGS than in MCD.245 CD80 also func-tions as an inhibitorymolecule in T cell–DC interaction and is downregulated byCTLA4, which is decreased in both se-rum and urine in active MCD.244 Thus,an initiating event, or first hit, such as aninfectious process, might lead to activa-tion of podocyte CD80 by IL13 or TLR 3or 4 ligands leading to actin rearrange-ment and albuminuria with CD80 shed-ding in the urine.245–247 The second hitwould involve defective CD80 regula-tion by either Tregs or podocyte-derivedCTLA4.246 Podocyte overexpression ofangiopoeitin-like-4, which, like CD80, isincreased in serum and podocytes inMCD patients, induces a steroid-sensitiveMCD-like glomerular lesion with heavyproteinuria, suggesting a role for thismol-ecule in the podocyte response.235 Finally,recent studies also suggest a role for pari-etal epithelial cells in formation of scle-rotic lesions.248,249

Membranous NephropathyIdiopathic membranous nephropathyis a noninflammatory glomerular lesionwith exclusively subepithelial deposits ofIgG and complement and heavy pro-teinuria.250,251 Heymann nephritis is arat model that closely mimics the humandisease.252 Studies in both active andpassive Heymann nephritis modelsshow that IgG antibodies form subepi-thelial immune deposits in situ by bind-ing to a podocyte protein complex nowcalled megalin,18,19,253 and that protein-uria is mediated by sublytic C5b-9 attack

on podocytes.1,2,83 Sublytic C5b-9 acti-vates several signaling pathways, altersthe actin cytoskeleton, and upregulatesexpression of TGFb and TGFb receptorsand matrix production leading to GBMthickening and spike formation. In-creased podocyte production of oxidantsand proteases damages underlying GBMleading to proteinuria.83 C5b-9 alsoleads to podocyte DNA damage and im-paired ability to complete the cell cycle,which may contribute to apoptosis, po-docytopenia, shedding of podocytes inthe urine, and development of glomeru-lar sclerosis.254

Proof of principle that membranousnephropathy in humans can also resultfrom an analogous autoimmune mecha-nism was first provided by Debiec et al.,who reported alloimmunization of an in-fant to neutral endopeptidase (NEP) ex-pressed on podocytes, resulting from amaternal NEP deficiency, which led totransplacental transfer of anti-NEP IgGand typical membranous nephropathy inthe newborn.255 However, the anti-NEPmechanism is not operative in most casesof adult idiopathic membranous ne-phropathy.256 Recently, Beck et al., usingmicrodissection and proteomic technol-ogy, identified another antipodocyte auto-antibody directed against the M-typephospholipase A2 receptor (PLA2R) in70%–80%of patients with primarymem-branous nephropathy and showed thatIgG anti-PLA2R was present in the glo-merular deposits and correlated withdisease activity, response to therapy, andrecurrence in transplants.257,258 Othershave confirmed these findings.259,260 An-tibodies reactive with aldose reductaseenolase and SOD as well as PLA2R havealso been eluted from membranous glo-meruli, but thesemay represent secondaryphenomena related to oxidant stress ratherthan primary pathogenic mediators.259

Whether the role of C5b-9 in medi-ating proteinuria as established in Hey-mann nephritis, and in the chronicserum sickness models of membranousnephropathy as well,261,262 mediates po-docyte injury and proteinuria in humanmembranous nephropathy is unclear.Complement-independent mechanismsof proteinuria are also well described

with IgG antipodocyte antibodies in sev-eral models,263–265 including Heymannnephritis,266,267 although these modelsdo not exhibit the prominent C3 andC5b-9 deposits seen in the complement-dependent Heymann models and inhumans.268,269 Despite the prominentcomplement deposition in membra-nous nephropathy, deposited anti-PLA2R antibody is predominately ofthe poorly complement-fixing IgG4 sub-class, although complement activationmight be induced by the lesser quantitiesof IgG1 and IgG3 usually present as occurswith anti-NEP IgG.256 However, in bothhuman membranous nephropathy andHeymann nephritis, classic complementpathway components are often absent inglomerulardeposits.269 Preliminary studieshave reported that IgG4 anti-PLA2Rbound to podocytes can activate C via theMBL pathway and induce sublytic podo-cyte injury analogous to the mechanismsdefined in the Heymannmodels in rats.270

Membranous nephropathy can spon-taneously remit,271 but once developed,the glomerular lesion heals very slowlyresulting in persistent proteinuria forweeks or months after the immune re-sponse has abated and subepithelialdeposits no longer are forming.272 Thislikely explains why only 70%–80%of pa-tients with proteinuria and membra-nous nephropathy on biopsy have activedisease as defined by elevated anti-PLAR2 levels.257,258 Glomerular deposi-tion of C3c and urinary excretion ofC5b-9 have both been established exper-imentally as valid biomarkers of ongoingimmune deposit formation in membra-nous nephropathy,272,273 but theseshould soon be supplanted by directmeasurements of anti-PLA2R antibodyin serum,274 which correlates with dis-ease activity and response to therapy.275

Although a role for cytotoxic T cells isproposed in complement-independentmodels of Heymann nephritis,276,277 Tcells do not have access to podocytes orthe subepithelial space and are rarely seenin most Heymann nephritis models orhuman membranous nephropathy.269,278

No systematic studies of the role of T cellsin human membranous nephropathyhave been reported.

10 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 11: Basic and Translational Concepts of Immune-Mediated Glomerular

Noetiologicagentshavebeen identifiedconsistently in idiopathic membranousnephropathy. However, a genome-wideassociation study reports very strongassociations with single nucleotide poly-morphisms in genes that encode forHLA-DQA1 and PLA2R.279 Whetherthese associations relate to renderingPLA2R antigenic or to altering its ex-pression by podocytes is unclear.

Anumberof potential etiologic agentshave been identified in secondary formsof membranous nephropathy, includinghepatitis B and C virus infection, severaldrugs, exposure to environmental toxinssuch as hydrocarbons, formaldehyde, cat-ionic BSA in cows’ milk (in infants),280

and solid organ tumors.269 Although30% of patients with tumor-associatedmembranous were found to be positivefor anti-PLA2R in one study,281 these sec-ondary forms of membranous nephropa-thy, including class V lupus membranousnephropathy, have generally not been as-sociatedwith anti-PLA2R, and their path-ogenesis remains unknown.

C3 GlomerulopathiesDense Deposit DiseaseDense deposit disease (DDD) has beenreferred to as type II MPGN because aminority of cases resemble MPGN I bylight microscopy and can have a similarnephritic/nephrotic clinical presenta-tion.199,282–284 However, DDD lacks Igdeposits and has little pathogenic over-lap with adult MPGN I, which is an im-mune complex disease. DDD is now bestviewed as a C3 glomerulopathy, a formof GN characterized by deposits of com-plement without Igs and usually associ-ated with abnormalities in complementregulation.285,286 DDD is a disorder ofalternative complement pathway regula-tion characterized by linear depositionof alternative pathway and terminalcomplement proteins including C5b-9,without IgG, along the contours ofribbon-like intramembranous electron-dense deposits within GBM and in themesangium (mesangial rings).282–284,287

The complement profile in serum and inglomerular deposits reflects alternative, orMBL, pathway activation.7–10,198,199,282–284

Normally, low-level spontaneous

hydrolysis of C3 to produce C3b leadsto formation of the alternative pathwayC3 convertase, C3bBb,which then cata-lyzes more C3 activation (C3 tickover).C3bBb is tightly regulated by circulatingcomplement factor H (CFH), whichbinds the active Bb site on the convertaseto impair degradation of the enzymeand prolong its half-life, leading to hy-percatabolism of C3.7–10,198,282–284,288

Over 80% of DDD patients have an IgGautoantibody to the Bb active site of thealternative pathway C3 convertase (C3nephritic factor, C3Nef) exposed afterinteraction of factor B with C3b thatprevents normal CFH binding.282–284

However, DDD can also be associatedwith other loss of CFH function condi-tions independent of C3Nef, includingcongenital absence or single nucleotidepolymorphisms of CFH, neutralizationby an anti-CFH antibody288–290 or anti-body to factor B,291 and, less commonly,with gain in function mutations in C3that lead to C3 convertases resistant toCFH regulation.284 In mice, CFH defi-ciency induces massive complement ac-tivation and a DDD phenotype that isameliorated by administration of CFHor properdin.292,293 Chronic unregu-lated C3 activation generates a varietyof alternative pathway complement acti-vation products that accumulate, per-haps by charge interactions, along theinner GBM to form the classic densedeposits seen by electron micro-scopy.284,287,290 In turn, this accumula-tion of proteinsmodifies filtration barrierstructure and integrity, leading to pro-teinuria and nephrotic syndrome. DDDis associated with other similar disordersof complement regulation such as par-tial lipodystrophy, but no specific etio-logic factors have been identified.

C3 NephropathyGlomerular deposits of C3 without Igalso characterize another C3 glomeru-lopathy variant, sometimes termed C3nephropathy or C3 deposition glomeru-lopathy; however, the electron-dense de-posits are primarily at mesangial andsubendothelial sites rather than withinGBM.7,285,286 These lesions also may beassociated with a spectrum of histologic

abnormalities including MPGN I–likefindings.285,286 The disorder seems toaffect younger patients who often havehematuria and proteinuria but less com-monly exhibit hypocomplementemia,nephrotic syndrome, or progression com-pared with DDD.288 Evidence of disor-dered complement regulation in theform of either mutated CRPs (H402 alleleof factor H, factor I), anti-CFH or anti-factor B antibodies, or C3Nef is also pres-ent in most of these patients.285,286,288–291

A familial form of the disease in peopleof Cypriot origin due to mutations inCFHR (CFHR5nephropathy)has recentlybeen described.294 The composition of thedeposits and the reason for their differentdistribution compared with DDD are notknown, although studies in murine mod-els of MPGN suggest that abnormalitiesin complement factor I may play a role.295

OVERVIEW OF IMMUNEMECHANISMS

Recent advances in understanding thepathogenesis of immune glomerulardiseases now link infectious processes,especially chronic viral ones, with auto-immunity andGN.Although once viewedprimarily as human equivalents of theantibody-mediated serumsickness (IC)orNTN (anti-GBM) models of GN in ani-mals, most human glomerulonephritidesare now believed to be primarily autoim-mune diseases. They involve both innateand adaptive immune mechanisms, withdistinction between the two becomingincreasingly blurred, andT cell aswell asantibody-driven adaptive immune re-sponses (Figures 1 through 4). Links toetiologic infectious agents more likelyproceed through recognition of PAMPsby TLRs and triggering of autoimmuneprocesses than through direct effects ofICs containing exogenous antigens trappedfrom the circulation.However, progress intranslating these scientific advances tobetter therapies has been slow, and clini-cians currently still rely almost entirely oncorticosteroids and toxic, nonselectiveimmunosuppressive agents for treatment.

As the relevant sciences have evolved,three things have remained constant—the

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 11

www.jasn.org BRIEF REVIEW

Page 12: Basic and Translational Concepts of Immune-Mediated Glomerular

patients, the utility of well character-ized animal models of their diseases, andthe contributions of physician-scientistswho have accounted for most of the ad-vances described above. The patients andthe animal models will remain and thetechnology to study them will advancedramatically in the years ahead. However,the future supply of qualified physician-scientists is threatened. In closing, it isworth noting that continued progress inthis area will require the continuous avail-ability anddedication of investigators whofully understand both the tools of basicscience and the clinical and pathologicmanifestations of human renal diseases,for knowledge of both will be requiredto generate and test new hypotheses thatcan lead to improvements in therapy. Per-haps this review can serve as an encourage-ment to some whomight follow that path.

ACKNOWLEDGMENTS

The author thanks Michael Carey, whose

willingness to undergo untested therapies for

his Goodpasture’s syndrome in 1966 ignited

my initial interest in immunologic glomer-

ular diseases; Edmund Lewis, who nurtured

and expanded that interest; and several for-

mer fellows and colleagues who have pro-

vided continuous intellectual and scientific

stimulation to understand more—particularly

David Salant, Kline Bolton, Rick Johnson, and

Masaomi Nangaku.

DISCLOSURESNone.

REFERENCES

1. Nangaku M, Couser WG: Mechanisms ofimmune-deposit formation and the media-tion of immune renal injury.Clin ExpNephrol9: 183–191, 2005

2. Ponticelli C, Coppo R, Salvadori M: Glo-merular diseases and transplantation: Simi-larities in pathogenetic mechanisms andtreatment options. Nephrol Dial Transplant26: 35–41, 2011

3. Anders HJ: Toll-like receptors and dangersignaling in kidney injury. J Am SocNephrol21: 1270–1274, 2010

4. Anders HJ, Muruve DA: The inflammasomesin kidney disease. J Am Soc Nephrol 22:1007–1018, 2011

5. Rosin DL, Okusa MD: Dangers within:DAMP responses to damage and cell deathin kidney disease. J Am Soc Nephrol 22:416–425, 2011

6. Gonçalves GM, Castoldi A, Braga TT,Câmara NO: New roles for innate immuneresponse in acute and chronic kidney in-juries. Scand J Immunol 73: 428–435, 2011

7. Pickering M, Cook HT: Complement andglomerular disease: New insights. Curr OpinNephrol Hypertens 20: 271–277, 2011

8. Puri TS, Quigg RJ: The many effects of com-plementC3- andC5-bindingproteins in renalinjury. Semin Nephrol 27: 321–337, 2007

9. Lesher AM, SongWC: Review: Complementand its regulatory proteins in kidney diseases.Nephrology (Carlton) 15: 663–675, 2010

10. Chen M, Daha MR, Kallenberg CG: Thecomplement system in systemic autoim-mune disease. J Autoimmun 34: J276–J286,2010

11. Daha NA, Banda NK, Roos A, Beurskens FJ,Bakker JM, Daha MR, Trouw LA: Comple-ment activation by (auto-) antibodies. MolImmunol 48: 1656–1665, 2011

12. Schroeder HW Jr, Cavacini L: Structure andfunction of immunoglobulins. J Allergy ClinImmunol 125[Suppl 2]: S41–S52, 2010

13. Couser WG, Baker PJ, Adler S: Comple-ment and the direct mediation of immuneglomerular injury: A new perspective. Kid-ney Int 28: 879–890, 1985

14. Stevens MG, Van Poucke M, Peelman LJ,Rainard P, De Spiegeleer B, Rogiers C, Vande Walle GR, Duchateau L, Burvenich C:Anaphylatoxin C5a-induced toll-like re-ceptor 4 signaling in bovine neutrophils.J Dairy Sci 94: 152–164, 2011

15. Dixon FJ, Feldman JD, Vazquez JJ: Experi-mental glomerulonephritis. The pathogen-esis of a laboratory model resembling thespectrum of human glomerulonephritis. J ExpMed 113: 899–920, 1961

16. Wilson CB, Dixon FJ: Diagnosis of immu-nopathologic renal disease. Kidney Int 5:389–401, 1974

17. Cochrane CG, Koffler D: Immune complexdisease in experimental animals and man.Adv Immunol 16: 185–264, 1973

18. Van Damme BJ, Fleuren GJ, Bakker WW,Vernier RL, Hoedemaeker PJ: Experimentalglomerulonephritis in the rat induced byantibodies directed against tubular anti-gens. V. Fixed glomerular antigens in thepathogenesis of heterologous immunecomplex glomerulonephritis. Lab Invest 38:502–510, 1978

19. Couser WG, Steinmuller DR, Stilmant MM,Salant DJ, Lowenstein LM: Experimentalglomerulonephritis in the isolated perfusedrat kidney. J Clin Invest 62: 1275–1287, 1978

20. Adler SG, Wang H, Ward HJ, Cohen AH,Border WA: Electrical charge. Its role in the

pathogenesis and prevention of experi-mental membranous nephropathy in therabbit. J Clin Invest 71: 487–499, 1983

21. Ward HJ, Cohen AH, Border WA: In situformation of subepithelial immune com-plexes in the rabbit glomerulus: Require-ment of a cationic antigen. Nephron 36:257–264, 1984

22. Couser WG, Salant DJ: In situ immunecomplex formation and glomerular injury.Kidney Int 17: 1–13, 1980

23. Couser WG: Mechanisms of glomerular in-jury in immune-complex disease. Kidney Int28: 569–583, 1985

24. Salant DJ, Adler S, Darby C, Capparell NJ,Groggel GC, Feintzeig ID, Rennke HG,Dittmer JE: Influence of antigen distributionon the mediation of immunological glomer-ular injury. Kidney Int 27: 938–950, 1985

25. Fries JW, Mendrick DL, Rennke HG: Deter-minants of immune complex-mediated glo-merulonephritis. Kidney Int 34: 333–345,1988

26. Wener MH, Mannik M: Mechanisms of im-mune deposit formation in renal glomeruli.Springer Semin Immunopathol 9: 219–235,1986

27. Strom TB, Koulmanda M: Recently dis-covered T cell subsets cannot keep theircommitments. J Am Soc Nephrol 20: 1677–1680, 2009

28. Rennke HG, Klein PS, Sandstrom DJ,Mendrick DL: Cell-mediated immune injuryin the kidney: Acute nephritis induced in therat by azobenzenearsonate. Kidney Int 45:1044–1056, 1994

29. Wu J, Hicks J, Borillo J, Glass WF 2nd, LouYH: CD4(+) T cells specific to a glomerularbasement membrane antigen mediate glo-merulonephritis. JClin Invest 109: 517–524,2002

30. KimAH,MarkiewiczMA, ShawAS: New rolesrevealed for T cells and DCs in glomerulo-nephritis. J Clin Invest 119: 1074–1076, 2009

31. Miossec P, Korn T, Kuchroo VK: Interleukin-17 and type 17 helper T cells.N Engl J Med361: 888–898, 2009

32. Turner JE, Paust HJ, Steinmetz OM, PanzerU: The Th17 immune response in renal in-flammation. Kidney Int 77: 1070–1075,2010

33. Kitching AR, Holdsworth SR: The emer-gence of TH17 cells as effectors of renalinjury. J AmSocNephrol 22: 235–238, 2011

34. Steinmetz OM, Summers SA, Gan PY,Semple T, Holdsworth SR, Kitching AR: TheTh17-defining transcription factor RORgtpromotes glomerulonephritis. J Am SocNephrol 22: 472–483, 2011

35. Turner JE, Paust HJ, Steinmetz OM, PetersA, Riedel JH, Erhardt A, Wegscheid C,Velden J, Fehr S, Mittrücker HW, Tiegs G,Stahl RA, Panzer U: CCR6 recruits regulatoryT cells and Th17 cells to the kidney in glo-merulonephritis. J Am Soc Nephrol 21:974–985, 2010

12 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 13: Basic and Translational Concepts of Immune-Mediated Glomerular

36. Abdulahad WH, Stegeman CA, KallenbergCG: Review article: The role of CD4(+) Tcells in ANCA-associated systemic vasculi-tis. Nephrology (Carlton) 14: 26–32, 2009

37. Rodriguez-Iturbe B, Musser JM: The currentstate of poststreptococcal glomerulonephri-tis. J Am Soc Nephrol 19: 1855–1864, 2008

38. Rodríguez-Iturbe B, Batsford S: Pathogenesisof poststreptococcal glomerulonephritis acentury afterClemens von Pirquet.Kidney Int71: 1094–1104, 2007

39. Eison TM, Ault BH, Jones DP, Chesney RW,Wyatt RJ: Post-streptococcal acute glo-merulonephritis in children: Clinical fea-tures and pathogenesis. Pediatr Nephrol26: 165–180, 2011

40. Beres SB, Sesso R, Pinto SW, Hoe NP,Porcella SF, Deleo FR, Musser JM: Genomesequence of a Lancefield group C Strepto-coccus zooepidemicus strain causing epi-demic nephritis: New information about anold disease. PLoS ONE 3: e3026, 2008

41. Brodsky SV, Nadasdy T: Infection-relatedglomerulonephritis. Contrib Nephrol 169:153–160, 2011

42. Edelstein CL, Bates WD: Subtypes of acutepostinfectious glomerulonephritis: A clinico-pathological correlation. Clin Nephrol 38:311–317, 1992

43. Sorger K, Gessler U, Hübner FK, Köhler H,SchulzW,StühlingerW,ThoenesGH,ThoenesW: Subtypes of acute postinfectious glomer-ulonephritis. Synopsis of clinical and patho-logical features. Clin Nephrol 17: 114–128,1982

44. Luo YH, ChuangWJ, Wu JJ, Lin MT, Liu CC,Lin PY, Roan JN,Wong TW,Chen YL, Lin YS:Molecular mimicry between streptococcalpyrogenic exotoxin B and endothelial cells.Lab Invest 90: 1492–1506, 2010

45. Fujigaki Y, Nagase M, Kojima K, YamamotoT, Hishida A: Glomerular handling of im-mune complex in the acute phase of activein situ immune complex glomerulonephri-tis employing cationized ferritin in rats.Ultrastructural localization of immune com-plex, complements and inflammatory cells.Virchows Arch 431: 53–61, 1997

46. Fujigaki Y, Batsford SR, Bitter-Suermann D,Vogt A: Complement system promotes trans-fer of immune complex across glomerularfiltration barrier. Lab Invest 72: 25–33, 1995

47. Nordstrand A, Norgren M, Holm SE: Patho-genicmechanism of acute post-streptococcalglomerulonephritis. Scand J Infect Dis 31:523–537, 1999

48. Sesso RC, Ramos OL, Pereira AB: Detectionof IgG-rheumatoid factor in sera of patientswith acute poststreptococcal glomerulone-phritis and its relationship with circulatingimmunecomplexes. Clin Nephrol 26: 55–60, 1986

49. Vilches AR,WilliamsDG: Persistent anti-DNAantibodies and DNA-anti-DNA complexes inpost-streptococcal glomerulonephritis. ClinNephrol 22: 97–101, 1984

50. Ardiles LG, ValderramaG,Moya P,MezzanoSA: Incidence and studies on antigenicspecificities of antineutrophil-cytoplasmicautoantibodies (ANCA) in poststreptococcalglomerulonephritis. Clin Nephrol 47: 1–5,1997

51. Satoskar AA, Nadasdy G, Plaza JA, SedmakD, Shidham G, Hebert L, Nadasdy T: Staph-ylococcus infection-associated glomerulo-nephritis mimicking IgA nephropathy. Clin JAm Soc Nephrol 1: 1179–1186, 2006

52. Nasr SH, Markowitz GS, Stokes MB, SaidSM, Valeri AM, D’Agati VD: Acute post-infectious glomerulonephritis in the mod-ern era: Experience with 86 adults andreview of the literature. Medicine (Balti-more) 87: 21–32, 2008

53. D’AmicoG: Natural history of idiopathic IgAnephropathy and factors predictive of dis-ease outcome. Semin Nephrol 24: 179–196, 2004

54. Tumlin JA, Madaio MP, Hennigar R: Idio-pathic IgA nephropathy: Pathogenesis,histopathology, and therapeutic options.Clin J Am Soc Nephrol 2: 1054–1061, 2007

55. Coppo R, Feehally J, Glassock RJ: IgA ne-phropathy at two score and one. Kidney Int77: 181–186, 2010

56. Floege J: The pathogenesis of IgA ne-phropathy: What is new and how does itchange therapeutic approaches? Am J Kid-ney Dis 58: 992–1004, 2011

57. Suzuki H, Kiryluk K, Novak J, Moldoveanu Z,Herr AB, Renfrow MB, Wyatt RJ, Scolari F,Mestecky J, Gharavi AG, Julian BA: Thepathophysiology of IgA nephropathy. J AmSoc Nephrol 22: 1795–1803, 2011

58. Mestecky J, Tomana M, Crowley-NowickPA, Moldoveanu Z, Julian BA, Jackson S:Defective galactosylation and clearance ofIgA1 molecules as a possible etiopatho-genic factor in IgA nephropathy. ContribNephrol 104: 172–182, 1993

59. Novak J, Julian BA, TomanaM, Mestecky J:IgA glycosylation and IgA immune com-plexes in the pathogenesis of IgA ne-phropathy. Semin Nephrol 28: 78–87, 2008

60. Kiryluk K, Moldoveanu Z, Sanders JT, EisonTM, Suzuki H, Julian BA, Novak J, GharaviAG, Wyatt RJ: Aberrant glycosylation ofIgA1 is inherited in both pediatric IgA ne-phropathy and Henoch-Schönlein purpuranephritis. Kidney Int 80: 79–87, 2011

61. Yu HH, Chu KH, Yang YH, Lee JH,Wang LC,Lin YT, Chiang BL: Genetics and im-munopathogenesis of IgA nephropathy.ClinRev Allergy Immunol 41: 198–213, 2011

62. Smith AC, de Wolff JF, Molyneux K,Feehally J, Barratt J: O-glycosylation ofserum IgD in IgA nephropathy. J Am SocNephrol 17: 1192–1199, 2006

63. Suzuki Y, Suzuki H, Nakata J, Sato D,Kajiyama T, Watanabe T, Tomino Y: Patho-logical role of tonsillar B cells in IgA ne-phropathy. Clin Dev Immunol 2011: 639074,2011

64. Barratt J, Eitner F, Feehally J, Floege J:Immune complex formation in IgA ne-phropathy: A case of the ‘right’ antibodiesin the ‘wrong’ place at the ‘wrong’ time?Nephrol Dial Transplant 24: 3620–3623,2009

65. Kokubo T, Hiki Y, Iwase H, Tanaka A, TomaK, Hotta K, Kobayashi Y: Protective role ofIgA1 glycans against IgA1 self-aggregationand adhesion to extracellular matrix proteins.J Am Soc Nephrol 9: 2048–2054, 1998

66. Suzuki H, Kiryluk K, Novak J, Moldoveanu Z,Herr AB, Renfrow MB, Wyatt RJ, Scolari F,Mestecky J, Gharavi AG, Julian BA: Thepathophysiology of IgA nephropathy. J AmSoc Nephrol 22: 1795–1805, 2011

67. Wang Y, Zhao MH, Zhang YK, Li XM, WangHY: Binding capacity and pathophysiologi-cal effects of IgA1 from patients with IgAnephropathy on human glomerular me-sangial cells. Clin Exp Immunol 136: 168–175, 2004

68. Ballardie FW, Brenchley PE, Williams S,O’Donoghue DJ: Autoimmunity in IgA ne-phropathy. Lancet 2: 588–592, 1988

69. Suzuki H, Fan R, Zhang Z, Brown R, Hall S,Julian BA, Chatham WW, Suzuki Y, WyattRJ, Moldoveanu Z, Lee JY, Robinson J,TomanaM, Tomino Y, Mestecky J, Novak J:Aberrantly glycosylated IgA1 in IgA ne-phropathy patients is recognized by IgGantibodies with restricted heterogeneity.J Clin Invest 119: 1668–1677, 2009

70. Mauer SM, Sutherland DE, Howard RJ, FishAJ, Najarian JS,Michael AF: The glomerularmesangium. 3. Acute immune mesangialinjury: A new model of glomerulonephritis.J Exp Med 137: 553–570, 1973

71. Yamamoto T, Wilson CB: Quantitative andqualitative studies of antibody-inducedmesangial cell damage in the rat. Kidney Int32: 514–525, 1987

72. Yamamoto T, Wilson CB: Complement de-pendence of antibody-induced mesangialcell injury in the rat. J Immunol 138: 3758–3765, 1987

73. Brandt J, Pippin J, Schulze M, Hänsch GM,Alpers CE, Johnson RJ, Gordon K, CouserWG: Role of the complement membraneattack complex (C5b-9) in mediating ex-perimental mesangioproliferative glomer-ulonephritis. Kidney Int 49: 335–343, 1996

74. FloegeJ, JohnsonRJ,CouserWG:Mesangialcells in the pathogenesis of progressive glo-merular disease in animal models. Clin In-vestig 70: 857–864, 1992

75. Emancipator SN: Prospects and perspectiveson IgA nephropathy from animal models.Contrib Nephrol 169: 126–152, 2011

76. Johnson RJ: The glomerular response toinjury: Progression or resolution? Kidney Int45: 1769–1782, 1994

77. Moura IC, BenhamouM, Launay P, VrtovsnikF, Blank U, Monteiro RC: The glomerular re-sponse to IgA deposition in IgA nephropa-thy. Semin Nephrol 28: 88–95, 2008

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 13

www.jasn.org BRIEF REVIEW

Page 14: Basic and Translational Concepts of Immune-Mediated Glomerular

78. Monteiro RC: Role of IgA and IgA fc re-ceptors in inflammation. J Clin Immunol 30:1–9, 2010

79. Schlöndorff D, Banas B: The mesangial cellrevisited: No cell is an island. J Am SocNephrol 20: 1179–1187, 2009

80. Coppo R, Amore A, Peruzzi L, Vergano L,Camilla R: Innate immunity and IgA ne-phropathy. J Nephrol 23: 626–632, 2010

81. Rauterberg EW, Lieberknecht HM, WingenAM, Ritz E: Complement membrane attack(MAC) in idiopathic IgA-glomerulonephritis.Kidney Int 31: 820–829, 1987

82. Oortwijn BD, Eijgenraam JW, Rastaldi MP,Roos A, Daha MR, van Kooten C: The role ofsecretory IgA and complement in IgA ne-phropathy. Semin Nephrol 28: 58–65, 2008

83. Cybulsky AV: Membranous nephropathy.Contrib Nephrol 169: 107–125, 2011

84. Stangou M, Alexopoulos E, Pantzaki A,Leonstini M, Memmos D: C5b-9 glomerulardeposition and tubular alpha3beta1-integrinexpression are implicated in the develop-ment of chronic lesions and predict renalfunction outcome in immunoglobulin A ne-phropathy. Scand J Urol Nephrol 42: 373–380, 2008

85. EspinosaM,Ortega R,Gómez-Carrasco JM,López-Rubio F, López-Andreu M, López-Oliva MO, Aljama P: Mesangial C4d de-position: A new prognostic factor in IgAnephropathy. Nephrol Dial Transplant 24:886–891, 2009

86. Roos A, Rastaldi MP, Calvaresi N, OortwijnBD, Schlagwein N, van Gijlswijk-JanssenDJ, Stahl GL, Matsushita M, Fujita T, vanKooten C, Daha MR: Glomerular activationof the lectin pathway of complement in IgAnephropathy is associatedwithmore severerenal disease. J Am Soc Nephrol 17: 1724–1734, 2006

87. Sanders JS, Rutgers A, Stegeman CA,Kallenberg CG: Pulmonary-renal syndromewith a focus on anti-GBM disease. SeminRespir Crit Care Med 32: 328–334, 2011

88. Hudson BG, Tryggvason K, SundaramoorthyM, Neilson EG: Alport’s syndrome, Good-pasture’s syndrome, and type IV collagen.N Engl J Med 348: 2543–2556, 2003

89. Cochrane CG, Unanue ER, Dixon FJ: A roleof polymorphonuclear leukocytes and com-plement in nephrotoxic nephritis. J Exp Med122: 99–116, 1965

90. Lerner RA, Glassock RJ, Dixon FJ: The roleof anti-glomerular basement membraneantibody in the pathogenesis of humanglomerulonephritis. J Exp Med 126: 989–1004, 1967

91. Hébert MJ, Takano T, Papayianni A, RennkeHG, Minto A, Salant DJ, Carroll MC, BradyHR: Acute nephrotoxic serum nephritis incomplement knockout mice: Relative rolesof the classical and alternate pathways inneutrophil recruitment and proteinuria.Nephrol Dial Transplant 13: 2799–2803,1998

92. SheerinNS, Springall T,CarrollMC,HartleyB,Sacks SH: Protection against anti-glomerularbasement membrane (GBM)-mediated ne-phritis in C3- and C4-deficient mice.Clin ExpImmunol 110: 403–409, 1997

93. Zhao J, Yan Y, Cui Z, Yang R, Zhao MH: Theimmunoglobulin G subclass distribution ofanti-GBM autoantibodies against rHalpha3(IV)NC1 is associated with disease severity.Hum Immunol 70: 425–429, 2009

94. Cui Z, Zhao MH: Advances in human anti-glomerular basement membrane disease.Nat Rev Nephrol 7: 697–705, 2011

95. Cui Z, Wang HY, Zhao MH: Natural auto-antibodies against glomerular basementmembrane exist in normal human sera.Kidney Int 69: 894–899, 2006

96. Pedchenko V, Bondar O, Fogo AB, VanacoreR, Voziyan P, Kitching AR, Wieslander J,Kashtan C, Borza DB, Neilson EG, WilsonCB, Hudson BG: Molecular architecture ofthe Goodpasture autoantigen in anti-GBMnephritis. N Engl J Med 363: 343–354,2010

97. Hudson BG: The molecular basis of Good-pasture and Alport syndromes: Beacons forthe discovery of the collagen IV family. J AmSoc Nephrol 15: 2514–2527, 2004

98. Kalluri R, Cantley LG, Kerjaschki D, NeilsonEG: Reactive oxygen species expose cryp-tic epitopes associated with autoimmuneGoodpasture syndrome. J Biol Chem 275:20027–20032, 2000

99. Bolton WK, Chen L, Hellmark T, Fox J,Wieslander J: Molecular mapping of theGoodpasture’s epitope for glomerulone-phritis. Trans Am Clin Climatol Assoc 116:229–236, discussion 237–238, 2005

100. BoltonWK, Chen L, Hellmark T, WieslanderJ, Fox JW: Epitope spreading and autoim-mune glomerulonephritis in rats inducedby a T cell epitopeofGoodpasture’s antigen.J Am Soc Nephrol 16: 2657–2666, 2005

101. Rocklin RE, Lewis EJ, David JR: In vitro evi-dence for cellular hypersensitivity to glo-merular-basement-membrane antigens inhuman glomerulonephritis. N Engl J Med283: 497–501, 1970

102. Dixon FJ: What are sensitized cells doing inglomerulonephritis? N Engl J Med 283:536–537, 1970

103. Merkel F, Kalluri R, Marx M, Enders U,Stevanovic S, Giegerich G, Neilson EG,Rammensee HG, Hudson BG, Weber M:Autoreactive T-cells in Goodpasture’s syn-drome recognize the N-terminal NC1 do-main on alpha 3 type IV collagen. Kidney Int49: 1127–1133, 1996

104. Bolton WK, Tucker FL, Sturgill BC: Newavian model of experimental glomerulone-phritis consistent with mediation by cellularimmunity. Nonhumorally mediated glo-merulonephritis in chickens. J Clin Invest73: 1263–1276, 1984

105. Bolton WK, Chandra M, Tyson TM,Kirkpatrick PR, Sadovnic MJ, Sturgill BC:

Transfer of experimental glomerulonephri-tis in chickens bymononuclear cells. KidneyInt 34: 598–610, 1988

106. Tipping PG, Holdsworth SR: T cells in cres-centic glomerulonephritis. JAmSocNephrol17: 1253–1263, 2006

107. Couser WG: Pathogenesis of glomerulone-phritis: From chickens to humans. NephronPhysiol 112: 17–23, 2009

108. Ooi JD, Phoon RK, Holdsworth SR, KitchingAR: IL-23, not IL-12, directs autoimmunityto the Goodpasture antigen. J Am SocNephrol 20: 980–989, 2009

109. Phelps RG, Rees AJ: The HLA complex inGoodpasture’s disease: A model for ana-lyzing susceptibility to autoimmunity. Kid-ney Int 56: 1638–1653, 1999

110. Salama AD, Chaudhry AN, Holthaus KA,Mosley K, Kalluri R, Sayegh MH, LechlerRI, Pusey CD, Lightstone L: Regulation byCD25+ lymphocytes of autoantigen-specificT-cell responses in Goodpasture’s (anti-GBM) disease. Kidney Int 64: 1685–1694,2003

111. Olson SW, Arbogast CB, Baker TP,Owshalimpur D, Oliver DK, Abbott KC,Yuan CM: Asymptomatic autoantibodiesassociate with future anti-glomerular base-ment membrane disease. J Am Soc Nephrol22: 1946–1952, 2011

112. Arends J, Wu J, Borillo J, Troung L, Zhou C,Vigneswaran N, Lou YH: T cell epitope mim-icry in antiglomerular basement membranedisease. J Immunol 176: 1252–1258, 2006

113. Macconi D, Chiabrando C, Schiarea S,Aiello S, Cassis L, Gagliardini E, Noris M,Buelli S, Zoja C, Corna D, Mele C, Fanelli R,Remuzzi G, Benigni A: Proteasomal pro-cessing of albumin by renal dendritic cellsgenerates antigenic peptides. J Am SocNephrol 20: 123–130, 2009

114. Heymann F, Meyer-Schwesinger C,Hamilton-Williams EE, Hammerich L, PanzerU, Kaden S, Quaggin SE, Floege J, GröneHJ, Kurts C: Kidney dendritic cell activationis required for progression of renal diseasein amousemodel of glomerular injury. J ClinInvest 119: 1286–1297, 2009

115. Hochheiser K, Engel DR, Hammerich L,Heymann F, Knolle PA, Panzer U, Kurts C:Kidney dendritic cells become pathogenicduring crescentic glomerulonephritis withproteinuria. J Am Soc Nephrol 22: 306–316, 2011

116. Sung SS, Bolton WK: T cells and dendriticcells in glomerular disease: The new glo-merulotubular feedback loop. Kidney Int77: 393–399, 2010

117. Lindic J, Vizjak A, Ferluga D, Kovac D, Ales A,KvederR,PonikvarR,BrenA:Clinical outcomeof patients with coexistent antineutrophil cy-toplasmic antibodies and antibodies againstglomerular basementmembrane. Ther ApherDial 13: 278–281, 2009

118. Hecht N, Omoloja A, Witte D, CanessaL: Evolution of antiglomerular basement

14 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 15: Basic and Translational Concepts of Immune-Mediated Glomerular

membrane glomerulonephritis into membra-nous glomerulonephritis. Pediatr Nephrol23: 477–480, 2008

119. Stilmant MM, Bolton WK, Sturgill BC,Schmitt GW, Couser WG: Crescentic glo-merulonephritis without immune deposits:Clinicopathologic features. Kidney Int 15:184–195, 1979

120. Falk RJ, Jennette JC: Anti-neutrophil cyto-plasmic autoantibodies with specificity formyeloperoxidase in patients with systemicvasculitis and idiopathic necrotizing andcrescentic glomerulonephritis. N Engl JMed 318: 1651–1657, 1988

121. BerdenAE, Ferrario F, Hagen EC, JayneDR,Jennette JC, Joh K, Neumann I, Noël LH,Pusey CD, Waldherr R, Bruijn JA, BajemaIM: Histopathologic classification of ANCA-associated glomerulonephritis. J Am SocNephrol 21: 1628–1636, 2010

122. Rutgers A, Sanders JS, Stegeman CA,Kallenberg CG: Pauci-immune necrotizingglomerulonephritis. Rheum Dis Clin NorthAm 36: 559–572, 2010

123. Falk RJ, Gross WL, Guillevin L, Hoffman G,Jayne DR, Jennette JC, Kallenberg CG,Luqmani R, Mahr AD, Matteson EL, MerkelPA, Specks U, Watts R: Granulomatosis withpolyangiitis (Wegener’s): Analternativenamefor Wegener’s granulomatosis. J Am SocNephrol 22: 587–588, 2011

124. Falk RJ, Jennette JC: ANCAdisease:Whereis this field heading? J Am Soc Nephrol 21:745–752, 2010

125. Jennette JC, Xiao H, Falk R, Gasim AM:Experimental models of vasculitis and glo-merulonephritis induced by antineutrophilcytoplasmic autoantibodies.ContribNephrol169: 211–220, 2011

126. Chen M, Kallenberg CG: ANCA-associatedvasculitides—advances in pathogenesis andtreatment. Nat Rev Rheumatol 6: 653–664,2010

127. Wilde B, van Paassen P, Witzke O, TervaertJW: New pathophysiological insights andtreatment of ANCA-associated vasculitis.Kidney Int 79: 599–612, 2011

128. Flint J, Morgan MD, Savage CO: Pathogen-esis of ANCA-associated vasculitis. RheumDis Clin North Am 36: 463–477, 2010

129. Radford DJ, Savage CO, Nash GB: Treat-ment of rolling neutrophils with anti-neutrophil cytoplasmic antibodies causesconversion to firm integrin-mediated ad-hesion. Arthritis Rheum 43: 1337–1345,2000

130. Nagao T, Matsumura M, Mabuchi A, Ishida-Okawara A, Koshio O, Nakayama T,Minamitani H, Suzuki K: Up-regulation ofadhesion molecule expression in glomerularendothelial cells by anti-myeloperoxidaseantibody. Nephrol Dial Transplant 22: 77–87, 2007

131. van Rossum AP, Rarok AA, Huitema MG,Fassina G, Limburg PC, Kallenberg CG: Con-stitutive membrane expression of proteinase

3 (PR3) and neutrophil activation by anti-PR3 antibodies. J Leukoc Biol 76: 1162–1170, 2004

132. Falk RJ, Terrell RS, Charles LA, JennetteJC: Anti-neutrophil cytoplasmic autoanti-bodies induce neutrophils to degranulateand produce oxygen radicals in vitro. ProcNatl Acad Sci USA 87: 4115–4119, 1990

133. Kessenbrock K, KrumbholzM, SchönermarckU, Back W, Gross WL, Werb Z, Gröne HJ,Brinkmann V, Jenne DE: Netting neutrophilsin autoimmune small-vessel vasculitis. NatMed 15: 623–625, 2009

134. Bosch X: LAMPs and NETs in the patho-genesis of ANCA vasculitis. J Am SocNephrol 20: 1654–1656, 2009

135. Jerke U, Rolle S, Dittmar G, Bayat B,Santoso S, Sporbert A, Luft F, Kettritz R:Complement receptor Mac-1 is an adaptorfor NB1 (CD177)-mediated PR3-ANCAneutrophil activation. J Biol Chem 286:7070–7081, 2011

136. Summers SA, Steinmetz OM, Gan PY, OoiJD, Odobasic D, Kitching AR, HoldsworthSR: Toll-like receptor 2 induces Th17 mye-loperoxidase autoimmunity while Toll-likereceptor 9 drives Th1 autoimmunity in mu-rine vasculitis. Arthritis Rheum 63: 1124–1135, 2011

137. Johnson RJ, Couser WG, Chi EY, Adler S,Klebanoff SJ: New mechanism for glo-merular injury. Myeloperoxidase-hydrogenperoxide-halide system. J Clin Invest 79:1379–1387, 1987

138. Xiao H, Heeringa P, Hu P, Liu Z, Zhao M,Aratani Y, Maeda N, Falk RJ, Jennette JC:Antineutrophil cytoplasmic autoantibodiesspecific for myeloperoxidase cause glo-merulonephritis and vasculitis in mice.J Clin Invest 110: 955–963, 2002

139. Primo VC, Marusic S, Franklin CC,Goldmann WH, Achaval CG, Smith RN,Arnaout MA, Nikolic B: Anti-PR3 immuneresponses induce segmental and necrotizingglomerulonephritis. Clin Exp Immunol 159:327–337, 2010

140. Schreiber A, Xiao H, Falk RJ, Jennette JC:Bone marrow-derived cells are sufficientand necessary targets to mediate glomer-ulonephritis and vasculitis induced by anti-myeloperoxidase antibodies. J Am SocNephrol 17: 3355–3364, 2006

141. Little MA, Smyth L, Salama AD, MukherjeeS, Smith J, Haskard D, Nourshargh S, CookHT, Pusey CD: Experimental autoimmunevasculitis: An animalmodel of anti-neutrophilcytoplasmic autoantibody-associated sys-temic vasculitis. Am J Pathol 174: 1212–1220, 2009

142. Ruth AJ, Kitching AR, Kwan RY, OdobasicD, Ooi JD, Timoshanko JR, Hickey MJ,Holdsworth SR: Anti-neutrophil cytoplas-mic antibodies and effector CD4+ cells playnonredundant roles in anti-myeloperoxidasecrescentic glomerulonephritis. J Am SocNephrol 17: 1940–1949, 2006

143. Xiao H, Heeringa P, Liu Z, Huugen D, Hu P,Maeda N, Falk RJ, Jennette JC: The roleof neutrophils in the induction of glomeru-lonephritis by anti-myeloperoxidase anti-bodies. Am J Pathol 167: 39–45, 2005

144. Xiao H, Schreiber A, Heeringa P, Falk RJ,Jennette JC: Alternative complement path-way in the pathogenesis of disease me-diated by anti-neutrophil cytoplasmicautoantibodies. Am J Pathol 170: 52–64,2007

145. HuugenD, vanEschA,XiaoH,Peutz-KootstraCJ, Buurman WA, Tervaert JW, Jennette JC,Heeringa P: Inhibition of complement factorC5 protects against anti-myeloperoxidaseantibody-mediated glomerulonephritis inmice. Kidney Int 71: 646–654, 2007

146. Van Timmeren MM, Chen M, Heeringa P:Review article: Pathogenic role of com-plement activation in anti-neutrophil cyto-plasmic auto-antibody-associated vasculitis.Nephrology (Carlton) 14: 16–25, 2009

147. XingGQ, ChenM, LiuG, Heeringa P, ZhangJJ, Zheng X, e J, Kallenberg CG, Zhao MH:Complement activation is involved in renaldamage in human antineutrophil cytoplas-mic autoantibody associated pauci-immunevasculitis. J Clin Immunol 29: 282–291, 2009

148. Kain R, Exner M, Brandes R, Ziebermayr R,CunninghamD, Alderson CA, Davidovits A,Raab I, Jahn R, Ashour O, Spitzauer S,Sunder-Plassmann G, Fukuda M, Klemm P,Rees AJ, Kerjaschki D:Molecular mimicry inpauci-immune focal necrotizing glomeru-lonephritis. Nat Med 14: 1088–1096, 2008

149. Pendergraft WF 3rd, Preston GA, Shah RR,Tropsha A, Carter CW Jr, Jennette JC, FalkRJ: Autoimmunity is triggered by cPR-3(105-201), a protein complementary to hu-man autoantigen proteinase-3. Nat Med10: 72–79, 2004

150. Hewins P, Belmonte F, Charles Jennette J,Falk RJ, Preston GA: Longitudinal studies ofpatients with ANCA vasculitis demonstrateconcurrent reactivity to complementary PR3protein segments cPR3m and cPR3C andwith no reactivity to cPR3N. Autoimmunity44: 98–106, 2011

151. Preston GA, Pendergraft WF 3rd, Falk RJ:New insights that link microbes with thegeneration of antineutrophil cytoplasmicautoantibodies: The theory of autoantigencomplementarity. Curr Opin Nephrol Hy-pertens 14: 217–222, 2005

152. Bautz DJ, Preston GA, Lionaki S, Hewins P,Wolberg AS, Yang JJ, Hogan SL, Chin H,Moll S, Jennette JC, Falk RJ: Antibodieswith dual reactivity to plasminogen andcomplementary PR3 in PR3-ANCA vasculi-tis. J Am Soc Nephrol 19: 2421–2429, 2008

153. Chen M, Kallenberg CG, Zhao MH: ANCA-negative pauci-immune crescentic glomer-ulonephritis. Nat Rev Nephrol 5: 313–318,2009

154. Bolton WK, Innes DJ Jr, Sturgill BC, KaiserDL: T-cells and macrophages in rapidly

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 15

www.jasn.org BRIEF REVIEW

Page 16: Basic and Translational Concepts of Immune-Mediated Glomerular

progressive glomerulonephritis: Clinicopath-ologic correlations. Kidney Int 32: 869–876,1987

155. Cunningham MA, Huang XR, Dowling JP,Tipping PG, Holdsworth SR: Prominence ofcell-mediated immunity effectors in “pauci-immune” glomerulonephritis. J Am SocNephrol 10: 499–506, 1999

156. Wilde B, Thewissen M, Damoiseaux J, vanPaassen P, Witzke O, Tervaert JW: T cellsin ANCA-associated vasculitis: What canwe learn from lesional versus circulating Tcells? Arthritis Res Ther 12: 204, 2010

157. Nogueira E, Hamour S, Sawant D,Henderson S, Mansfield N, Chavele KM,Pusey CD, Salama AD: Serum IL-17 andIL-23 levels and autoantigen-specific Th17cells are elevated in patients with ANCA-associated vasculitis. Nephrol Dial Trans-plant 25: 2209–2217, 2010

158. King WJ, Brooks CJ, Holder R, Hughes P,Adu D, Savage CO: T lymphocyte responsesto anti-neutrophil cytoplasmic autoantibody(ANCA) antigens are present in patientswith ANCA-associated systemic vasculitisand persist during disease remission. ClinExp Immunol 112: 539–546, 1998

159. Brouwer E, Stegeman CA, Huitema MG,Limburg PC, KallenbergCG: T cell reactivityto proteinase 3 and myeloperoxidase inpatients with Wegener’s granulomatosis(WG). Clin Exp Immunol 98: 448–453, 1994

160. Yang J, Bautz DJ, Lionaki S, Hogan SL, ChinH, Tisch RM, Schmitz JL, Pressler BM,Jennette JC, Falk RJ, Preston GA: ANCApatients have T cells responsive to com-plementary PR-3 antigen. Kidney Int 74:1159–1169, 2008

161. Gan PY, Steinmetz OM, Tan DS, O’SullivanKM, Ooi JD, Iwakura Y, Kitching AR,Holdsworth SR: Th17 cells promote auto-immune anti-myeloperoxidase glomerulone-phritis. J Am Soc Nephrol 21: 925–931, 2010

162. Chavele KM, Shukla D, Keteepe-Arachi T,Seidel JA, Fuchs D, Pusey CD, Salama AD:Regulationofmyeloperoxidase-specific T cellresponses during disease remission in anti-neutrophil cytoplasmic antibody-associatedvasculitis: The role of Treg cells and trypto-phan degradation.Arthritis Rheum 62: 1539–1548, 2010

163. Chiang CK, Inagi R: Glomerular diseases:Genetic causes and future therapeutics.Nat Rev Nephrol 6: 539–554, 2010

164. Cao Y, Schmitz JL, Yang J, Hogan SL, BunchD, Hu Y, Jennette CE, Berg EA, Arnett FC Jr,Jennette JC, Falk RJ, Preston GA: DRB1*15allele is a risk factor for PR3-ANCA diseasein African Americans. J Am Soc Nephrol 22:1161–1167, 2011

165. Rutgers A, Slot M, van Paassen P, van BredaVriesman P, Heeringa P, Tervaert JW: Co-existence of anti-glomerular basementmembrane antibodies and myeloperoxidase-ANCAs in crescentic glomerulonephritis.AmJ Kidney Dis 46: 253–262, 2005

166. Nasr SH, D’Agati VD, Park HR, Sterman PL,Goyzueta JD,Dressler RM,Hazlett SM, PursellRN,CaputoC,MarkowitzGS:Necrotizingandcrescentic lupus nephritis with antineutrophilcytoplasmic antibody seropositivity.Clin JAmSoc Nephrol 3: 682–690, 2008

167. Nasr SH, Said SM, Valeri AM, Stokes MB,Masani NN, D’Agati VD, Markowitz GS:Membranousglomerulonephritis with ANCA-associated necrotizing and crescentic glo-merulonephritis. Clin J Am Soc Nephrol 4:299–308, 2009

168. Markowitz GS, D’Agati VD: Classification oflupus nephritis. Curr Opin Nephrol Hyper-tens 18: 220–225, 2009

169. Bhinder S, Singh A, Majithia V: Membra-nous (class V) renal disease in systemic lupuserythematosus may be more common thanpreviously reported: Results of a 6-yearretrospective analysis. Am J Med Sci 339:230–232, 2010

170. Agmon-LevinN,MoscaM, PetriM,ShoenfeldY: Systemic lupus erythematosus: One dis-ease or many [published online ahead ofprint October 25, 2011]? Autoimmun Rev

171. Crispín JC, Liossis SN, Kis-Toth K, LiebermanLA, Kyttaris VC, Juang YT, Tsokos GC: Path-ogenesis of human systemic lupus erythem-atosus: Recent advances. Trends Mol Med16: 47–57, 2010

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

173. Davidson A, Aranow C: Lupus nephritis:Lessons from murine models. Nat RevRheumatol 6: 13–20, 2010

174. Mannik M, Merrill CE, Stamps LD, WenerMH: Multiple autoantibodies form the glo-merular immune deposits in patients withsystemic lupus erythematosus. J Rheumatol30: 1495–1504, 2003

175. Mielle JE, RekvigOP, Van Der Vlag J, FentonKA: Nephritoigenic antibodies bind in glo-meruli through interaction with exposedchromatin fragments and not with renalcross-reactive antigens. Autoimmunity 44:373–383, 2011

176. Raz E, Brezis M, Rosenmann E, Eilat D: Anti-DNA antibodies bind directly to renal anti-gens and induce kidney dysfunction in theisolated perfused rat kidney. J Immunol142: 3076–3082, 1989

177. Waldman M, Madaio MP: Pathogenic au-toantibodies in lupus nephritis. Lupus 14:19–24, 2005

178. Yanase K, Madaio MP: Nuclear localizinganti-DNA antibodies enter cells via caveoliand modulate expression of caveolin andp53. J Autoimmun 24: 145–151, 2005

179. 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

180. Martinez-ValleF, BaladaE,Ordi-Ros J,Bujan-Rivas S, Sellas-Fernandez A, Vilardell-Tarres

M: DNase1 activity in systemic lupus erythem-atosus patients with and without ne-phropathy. Rheumatol Int 30: 1601–1604,2010

181. Pawar RD, Patole PS, Ellwart A, Lech M,Segerer S, Schlondorff D, Anders HJ: Li-gands to nucleic acid-specific toll-like re-ceptors and the onset of lupus nephritis.J Am Soc Nephrol 17: 3365–3373, 2006

182. Smith KD: Lupus nephritis: Toll the trigger!J Am Soc Nephrol 17: 3273–3275, 2006

183. Shen Y, Chen XW, Sun CY, Dai M, Yan YC,Yang CD: Association between anti-beta2glycoprotein I antibodies and renal glo-merular C4d deposition in lupus nephritispatients with glomerular microthrombosis:A prospective study of 155 cases. Lupus 19:1195–1203, 2010

184. Pickering MC, Botto M: Are anti-C1q anti-bodies different from other SLE autoanti-bodies? Nat Rev Rheumatol 6: 490–493,2010

185. Charles N, Hardwick D, Daugas E, Illei GG,Rivera J: Basophils and the T helper 2 en-vironment can promote the developmentof lupus nephritis. Nat Med 16: 701–707,2010

186. Marston B, Looney RJ: Connective tissuediseases: Translating the effects of BAFF inSLE. Nat Rev Rheumatol 6: 503–504, 2010

187. Bao L, Quigg RJ: Complement in lupusnephritis: The good, the bad, and the un-known. Semin Nephrol 27: 69–80, 2007

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

189. 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

190. Lu L, KaliyaperumalA, BoumpasDT,Datta SK:Major peptide autoepitopes for nucleosome-specific T cells of human lupus. J Clin Invest104: 345–355, 1999

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

192. Sawla P, Hossain A, Hahn BH, Singh RP:Regulatory T cells in systemic lupus erythem-atosus. Role of peptide tolerance [publishedonline ahead of print October 7, 2011].Autoimmun Rev (2011), doi:10.1016/j.autrev.2011.09.008

193. Strickland FM, Richardson BC: Epigeneticsin human autoimmunity. Epigenetics inautoimmunity - DNAmethylation in systemiclupus erythematosus and beyond. Autoim-munity 41: 278–286, 2008

194. Vuong MT, Gunnarsson I, Lundberg S,Svenungsson E, Wramner L, Fernström A,Syvänen AC, Do LT, Jacobson SH, PadyukovL: Genetic risk factors in lupus nephritis and

16 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 17: Basic and Translational Concepts of Immune-Mediated Glomerular

IgA nephropathy—no support of an overlap.PLoS ONE 5: e10559, 2010

195. Kraft SW, Schwartz MM, Korbet SM, LewisEJ: Glomerular podocytopathy in patientswith systemic lupus erythematosus. J AmSoc Nephrol 16: 175–179, 2005

196. Smith KD, Alpers CE: Pathogenic mecha-nisms in membranoproliferative glomeru-lonephritis. Curr Opin Nephrol Hypertens14: 396–403, 2005

197. Alpers CE, Smith KD: Cryoglobulinemiaand renal disease. Curr Opin Nephrol Hy-pertens 17: 243–249, 2008

198. Alchi B, Jayne D: Membranoproliferativeglomerulonephritis. Pediatr Nephrol 25:1409–1418, 2010

199. Vernon KA, Pickering MC, Cook T: Experi-mental models of membranoproliferativeglomerulonephritis, including dense de-posit disease. Contrib Nephrol 169: 198–210, 2011

200. Coleman TH, Forristal J, Kosaka T, WestCD: Inherited complement componentdeficiencies in membranoproliferative glo-merulonephritis. Kidney Int 24: 681–690,1983

201. Sethi S, Zand L, Leung N, Smith RJ,Jevremonic D, Herrmann SS, Fervenza FC:Membranoproliferative glomerulonephritissecondary to monoclonal gammopathy.Clin J Am Soc Nephrol 5: 770–782, 2010

202. Johnson RJ, Gretch DR, Yamabe H, Hart J,Bacchi CE, Hartwell P, CouserWG, Corey L,Wener MH, Alpers CE, Willson R: Mem-branoproliferative glomerulonephritis as-sociated with hepatitis C virus infection. NEngl J Med 328: 465–470, 1993

203. Johnson RJ, Willson R, Yamabe H, CouserW, Alpers CE, Wener MH, Davis C, GretchDR: Renal manifestations of hepatitis Cvirus infection. Kidney Int 46: 1255–1263,1994

204. Charles ED, Dustin LB: Hepatitis C virus-induced cryoglobulinemia. Kidney Int 76:818–824, 2009

205. Cacoub P, Ghillani P, Revelen R, Thibault V,Calvez V, Charlotte F, Musset L, Youinou P,Piette JC:Anti-endothelial cell auto-antibodiesin hepatitis C virus mixed cryoglobulinemia.J Hepatol 31: 598–603, 1999

206. Saadoun D, Sadallah S, Trendelenburg M,Limal N, Sene D, Piette JC, Schifferli JA,Cacoub P: Anti-C1q antibodies in hepatitisC virus infection. Clin Exp Immunol 145:308–312, 2006

207. McMurray RW: Hepatitis C-associated au-toimmune disorders. Rheum Dis Clin NorthAm 24: 353–374, 1998

208. Sansonno D, Lauletta G, Nisi L, Gatti P,Pesola F, Pansini N, Dammacco F: Non-envelopedHCV core protein as constitutiveantigen of cold-precipitable immune com-plexes in type II mixed cryoglobulinaemia.Clin Exp Immunol 133: 275–282, 2003

209. Sansonno D, Gesualdo L, Manno C, SchenaFP, Dammacco F: Hepatitis C virus-related

proteins in kidney tissue from hepatitisC virus-infected patients with cryoglobuline-mic membranoproliferative glomerulone-phritis. Hepatology 25: 1237–1244, 1997

210. Sansonno D, Tucci FA, Ghebrehiwet B,Lauletta G, Peerschke EI, Conteduca V,Russi S, Gatti P, Sansonno L, Dammacco F:Role of the receptor for the globular domainof C1q protein in the pathogenesis ofhepatitis C virus-related cryoglobulin vas-cular damage. J Immunol 183: 6013–6020,2009

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

212. Robson MG: Toll-like receptors and renaldisease. Nephron, Exp Nephrol 113: e1–e7, 2009

213. Pawar RD, Patole PS, Zecher D, Segerer S,Kretzler M, Schlöndorff D, Anders HJ: Toll-like receptor-7modulates immune complexglomerulonephritis. J Am Soc Nephrol 17:141–149, 2006

214. Mühlfeld AS, Segerer S, Hudkins K, Farr AG,Bao L, Kraus D, Holers VM, Quigg RJ,AlpersCE: Overexpression of complementinhibitor Crry does not prevent cryoglobulin-associated membranoproliferative glomer-ulonephritis. Kidney Int 65: 1214–1223,2004

215. Zhang S, Audard V, Fan Q, Pawlak A, LangP, Sahali D: Immunopathogenesis of idio-pathic nephrotic syndrome.ContribNephrol169: 94–106, 2011

216. Waldman M, Crew RJ, Valeri A, Busch J,Stokes B, Markowitz G, D’Agati V, Appel G:Adult minimal-change disease: Clinical char-acteristics, treatment, and outcomes. ClinJ Am Soc Nephrol 2: 445–453, 2007

217. DAgati VD, Kaskel FJ, Falk RJ: Focal seg-mental glomerulosclerosis. N Engl J Med365: 2398–2411, 2011

218. Shalhoub RJ: Pathogenesis of lipoid ne-phrosis: A disorder of T-cell function. Lancet2: 556–560, 1974

219. Koyama A, Fujisaki M, Kobayashi M,Igarashi M, Narita M: A glomerular perme-ability factor produced by human T cellhybridomas. Kidney Int 40: 453–460, 1991

220. McCarthy ET, Sharma M, Savin VJ: Circu-lating permeability factors in idiopathicnephrotic syndrome and focal segmentalglomerulosclerosis. Clin J Am Soc Nephrol5: 2115–2121, 2010

221. Mauer SM, Hellerstein S, Cohn RA, SibleyRK, Vernier RL: Recurrence of steroid-responsive nephrotic syndrome after renaltransplantation. J Pediatr 95: 261–264,1979

222. Ali AA, Wilson E, Moorhead JF, Amlot P,Abdulla A, FernandoON,DormanA, SwenyP: Minimal-change glomerular nephritis.Normal kidneys in an abnormal environ-ment? Transplantation 58: 849–852, 1994

223. ReaR, SmithC,SandhuK,KwanJ, TomsonC:Successful transplant of a kidney with focal

segmental glomerulosclerosis.Nephrol DialTransplant 16: 416–417, 2001

224. Liu Z, Blattner SM, Tu Y, Tisherman R,WangJH, Rastaldi MP, Kretzler M, Wu C: Alpha-actinin-4 and CLP36 protein deficienciescontribute to podocyte defects in multiplehuman glomerulopathies. J Biol Chem 286:30795–30805, 2011

225. Freedman BI, Kopp JB, Langefeld CD,Genovese G, Friedman DJ, Nelson GW,Winkler CA, Bowden DW, Pollak MR: Theapolipoprotein L1 (APOL1) gene and non-diabetic nephropathy in African Americans.J Am Soc Nephrol 21: 1422–1426, 2010

226. Genovese G, Friedman DJ, Ross MD,Lecordier L, Uzureau P, Freedman BI,Bowden DW, Langefeld CD, Oleksyk TK,Uscinski Knob AL, Bernhardy AJ, Hicks PJ,Nelson GW, Vanhollebeke B, Winkler CA,Kopp JB, Pays E, Pollak MR: Association oftrypanolytic ApoL1 variants with kidneydisease in African Americans. Science 329:841–845, 2010

227. Friedman DJ, Kozlitina J, Genovese G,Prachi Jog P,, Pollak MR: APOL1 genotypeidentifies African Americans at high risk ofnon-diabetic kidney disease in the DallasHeart Study. J Am Soc Nephrol 22: 2129–2137, 2011

228. Kopp JB, Nelson GW, Sampath K, JohnsonRC, Genovese G, An P, Friedman D, BriggsW, Dart R, Korbet S, Mokrzycki MH, KimmelPL, Limou S, Ahuja TS, Berns JS, Fryc J,Simon EE, Smith MC, Trachtman H, MichelDM, Schelling JR, Vlahov D, Pollak M,Winkler CA: APOL1 genetic variants in focalsegmental glomerulosclerosis and HIV-associated nephropathy. J Am Soc Nephrol22: 2129–2137, 2011

229. Papeta N, Kiryluk K, Patel A, Sterken R,Kacak N, Snyder HJ, Imus PH, Mhatre AN,Lawani AK, Julian BA, Wyatt RJ, Novak J,Wyatt CM, Ross MJ, Winston JA, KlotmanME, Cohen DJ, Appel GB, D’Agati VD,Klotman PE, Gharavi AG: APOL1 variantsincrease risk for FSGS and HIVAN but notIgA nephropathy. J Am Soc Nephrol 22:1991–1996, 2011

230. Wei C, El Hindi S, Li J, Fornoni A, Goes N,Sageshima J, Maiguel D, Karumanchi SA,Yap HK, Saleem M, Zhang Q, Nikolic B,Chaudhuri A, Daftarian P, Salido E, TorresA, Salifu M, Sarwal MM, Schaefer F, MorathC, Schwenger V, Zeier M, Gupta V, Roth D,Rastaldi MP, Burke G, Ruiz P, Reiser J: Cir-culating urokinase receptor as a cause offocal segmental glomerulosclerosis. NatMed 17: 952–960, 2011

231. Welch GI, Saleem MA: Podocyte cytoskel-eton: Key to functioning glomerulus inhealth and disease.Nat Rev Nephrol 8: 14–21, 2012

232. Zhu L, Jiang R,Aoudjit L, JonesN, TakanoT:Activation of RhoA in podocytes inducesfocal segmental glomerulosclerosis. J AmSoc Nephrol 22: 1621–1630, 2011

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 17

www.jasn.org BRIEF REVIEW

Page 18: Basic and Translational Concepts of Immune-Mediated Glomerular

233. Sistani L, Dunér F, Udumala S, Hultenby K,Uhlen M, Betsholtz C, Tryggvason K,Wernerson A, Patrakka J: Pdlim2 is a novelactin-regulating protein of podocyte footprocesses. Kidney Int 80: 1045–1054, 2011

234. Fuchshofer R, Ullmann S, Zeilbeck LF,Baumann M, Junglas B, Tamm ER: Con-nective tissue growth factor modulates po-docyte actin cytoskeleton and extracellularmatrix synthesis and is induced in podo-cytes upon injury. Histochem Cell Biol 136:301–319, 2011

235. Clement LC,Avila-CasadoC,MacéC,Soria E,Bakker WW, Kersten S, Chugh SS: Podocyte-secreted angiopoietin-like-4 mediates pro-teinuria in glucocorticoid-sensitive nephroticsyndrome. Nat Med 17: 117–122, 2011

236. Tain YL, Chen TY, Yang KD: Implications ofserum TNF-beta and IL-13 in the treatmentresponse of childhood nephrotic syndrome.Cytokine 21: 155–159, 2003

237. Yap HK, Cheung W, Murugasu B, Sim SK,Seah CC, Jordan SC: Th1 and Th2 cytokinemRNA profiles in childhood nephrotic syn-drome: Evidence for increased IL-13 mRNAexpression in relapse. J Am Soc Nephrol10: 529–537, 1999

238. Parry RG, Gillespie KM, Mathieson PW:Effects of type 2 cytokines on glomerularepithelial cells. Exp Nephrol 9: 275–283,2001

239. Van Den Berg JG, Aten J, Chand MA,Claessen N, Dijkink L, Wijdenes J, Lakkis FG,Weening JJ: Interleukin-4 and interleukin-13 act on glomerular visceral epithelial cells.J Am Soc Nephrol 11: 413–422, 2000

240. Lai KW, Wei CL, Tan LK, Tan PH, ChiangGS, Lee CG, Jordan SC, Yap HK: Over-expression of interleukin-13 induces minimal-change-like nephropathy in rats. J Am SocNephrol 18: 1476–1485, 2007

241. Sellier-LeclercAL,DuvalA, RiveronS,MacherMA, Deschenes G, Loirat C, Verpont MC,Peuchmaur M, Ronco P, Monteiro RC,Haddad E: A humanized mouse model ofidiopathic nephrotic syndrome suggests apathogenic role for immature cells. J AmSocNephrol 18: 2732–2739, 2007

242. Reiser J, von Gersdorff G, Loos M, Oh J,Asanuma K, Giardino L, Rastaldi MP,Calvaresi N, Watanabe H, Schwarz K, FaulC, Kretzler M, Davidson A, Sugimoto H,Kalluri R, Sharpe AH, Kreidberg JA, MundelP: Induction of B7-1 in podocytes is asso-ciated with nephrotic syndrome. J Clin In-vest 113: 1390–1397, 2004

243. Reiser J, Mundel P: Danger signaling byglomerular podocytes defines a novelfunction of inducible B7-1 in the patho-genesis of nephrotic syndrome. J Am SocNephrol 15: 2246–2248, 2004

244. Garin EH, Mu W, Arthur JM, Rivard CJ,Araya CE, Shimada M, Johnson RJ: UrinaryCD80 is elevated inminimal changediseasebut not in focal segmental glomerulo-sclerosis. Kidney Int 78: 296–302, 2010

245. Navarro-Munoz M, Ibernon M, Perez V, AraJ, Espinal A, Lopez D, Bonet J, Romero R:Messenger RNA expression of B7-1 andNPHS1 in urinary sediment could be usefulto differentiate between minimal changedisease and focal segmental glomerulo-sclerosis in adult patients. Nephrol DialTransplant 26: 3914–3923, 2011

246. Shimada M, Araya C, Rivard C, Ishimoto T,Johnson RJ, Garin EH: Minimal changedisease: A “two-hit” podocyte immunedisorder? Pediatr Nephrol 26: 645–649,2011

247. Shimada M, Ishimoto T, Lee PY, LanaspaMA, Rivard CJ, Roncal-Jimenez CA, WymerDT, Yamabe H,Mathieson PW, SaleemMA,Garin EH, Johnson RJ: Toll-like receptor 3ligands induce CD80 expression in humanpodocytes via an NF-{kappa}B-dependentpathway [published online ahead of printMay 26, 2011].Nephrol Dial Transplant doi:10.1093/ndt/gfr271

248. 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 inthe formation of sclerotic lesions in focalsegmental glomerulosclerosis. J Am SocNephrol 22: 1262–1274, 2011

249. Ohse T, Vaughan MR, Kopp JB, Krofft RD,Marshall CB, ChangAM,Hudkins KL, AlpersCE, Pippin JW, Shankland SJ: De novo ex-pression of podocyte proteins in parietalepithelial cells during experimental glo-merular disease.Am J Physiol Renal Physiol298: F702–F711, 2010

250. Ronco P, Debiec H: Membranous glomer-ulopathy: The evolving story. Curr OpinNephrol Hypertens 19: 254–259, 2010

251. Ronco P,DebiecH: Antigen identification inmembranous nephropathy moves towardtargetedmonitoring and new therapy. J AmSoc Nephrol 21: 564–569, 2010

252. Kerjaschki D: Pathomechanisms and mo-lecular basis of membranous glomerulo-pathy. Lancet 364: 1194–1196, 2004

253. Couser WG, Nangaku M: Cellular and mo-lecular biology of membranous nephropa-thy. J Nephrol 19: 699–705, 2006

254. Pippin JW, Durvasula R, Petermann A,Hiromura K, Couser WG, Shankland SJ:DNAdamage is a novel response to sublyticcomplement C5b-9-induced injury in po-docytes. J Clin Invest 111: 877–885, 2003

255. Debiec H, Guigonis V, Mougenot B,Decobert F, Haymann JP, Bensman A,Deschênes G, Ronco PM: Antenatal mem-branous glomerulonephritis due to anti-neutral endopeptidase antibodies. N EnglJ Med 346: 2053–2060, 2002

256. Ronco P, Debiec H: Target antigens andnephritogenic antibodies in membranousnephropathy: Of rats and men. Semin Im-munopathol 29: 445–458, 2007

257. Beck LH Jr, Bonegio RG, Lambeau G, BeckDM, Powell DW, Cummins TD, Klein JB,

Salant DJ: M-type phospholipase A2 re-ceptor as target antigen in idiopathic mem-branous nephropathy. N Engl J Med 361:11–21, 2009

258. Hofstra JM, Beck LH Jr, Beck DM, WetzelsJF, Salant DJ: Anti-phospholipase A₂ re-ceptor antibodies correlate with clinical sta-tus in idiopathic membranous nephropathy.Clin J Am Soc Nephrol 6: 1286–1291, 2011

259. Prunotto M, Carnevali ML, Candiano G,Murtas C, Bruschi M, Corradini E, Trivelli A,Magnasco A, Petretto A, Santucci L, MatteiS, Gatti R, Scolari F, Kador P, Allegri L,Ghiggeri GM: Autoimmunity in membra-nous nephropathy targets aldose reductaseand SOD2. J Am Soc Nephrol 21: 507–519,2010

260. Debiec H, Ronco P: PLA2R autoantibodiesand PLA2R glomerular deposits in mem-branous nephropathy. N Engl J Med 364:689–690, 2011

261. Groggel GC, Adler S, Rennke HG, CouserWG, Salant DJ: Role of the terminal com-plement pathway in experimental mem-branous nephropathy in the rabbit. J ClinInvest 72: 1948–1957, 1983

262. Couser WG, Ochi RF, Baker PJ, Schulze M,Campbell C, Johnson RJ: C6 depletion re-duces proteinuria in experimental nephrop-athy induced by a nonglomerular antigen.J Am Soc Nephrol 2: 894–901, 1991

263. CouserWG, StilmantMM, JermanovichNB:Complement-independent nephrotoxic ne-phritis in the guinea pig. Kidney Int 11: 170–180, 1977

264. Salant DJ, Madaio MP, Adler S, StilmantMM, Couser WG: Altered glomerular per-meability induced by F(ab’)2 and Fab’ anti-bodies to rat renal tubular epithelial antigen.Kidney Int 21: 36–43, 1982

265. Salant DJ, Natori Y, Kawachi H: Glomerularinjury due to antibody alone. In: Immuno-logic Renal Diseases, edited byNeilson EG,Couser WG, 2nd Ed., Philadelphia, Lippin-cott Wilkins, and Williams, 2001, pp 347–366

266. Leenaerts PL, Hall BM, Van Damme BJ,Daha MR, Vanrenterghem YF: Active Hey-mann nephritis in complement componentC6 deficient rats.Kidney Int 47: 1604–1614,1995

267. Spicer ST, Tran GT, Killingsworth MC,Carter N, Power DA, Paizis K, Boyd R,Hodgkinson SJ, Hall BM: Induction of pas-sive Heymann nephritis in complementcomponent 6-deficient PVG rats. J Immunol179: 172–178, 2007

268. Fervenza FC, Sethi S, Specks U: Idiopathicmembranous nephropathy: Diagnosis andtreatment. Clin J Am Soc Nephrol 3: 905–919, 2008

269. Couser WG, Cattran D: Membranous ne-phropathy. In: Comprehensive Clinical Ne-phrology, edited by Johnson RJ, Feehally J,Floege J, London, Mosby International Pub-lishers, 2010, pp 248–259

18 Journal of the American Society of Nephrology J Am Soc Nephrol 23: ccc–ccc, 2012

BRIEF REVIEW www.jasn.org

Page 19: Basic and Translational Concepts of Immune-Mediated Glomerular

270. Ma H, Beck LH, Salant DJ: Membranousnephropathy-associated anti-phospholipaseA2 receptor IgG4 autoantibodies activatethe lectin complement pathway [abstract].J Am Soc Nephrol 22: 62A, 2011

271. Polanco N, Gutiérrez E, Covarsí A, Ariza F,Carreño A, Vigil A, Baltar J, Fernández-Fresnedo G, Martín C, Pons S, Lorenzo D,Bernis C, Arrizabalaga P, Fernández-JuárezG, Barrio V, Sierra M, Castellanos I, EspinosaM, Rivera F,Oliet A, Fernández-Vega F, PragaM Grupo de Estudio de las EnfermedadesGlomerulares de la Sociedad Española deNefrología: Spontaneous remission of ne-phrotic syndrome in idiopathic membranousnephropathy. J Am SocNephrol 21: 697–704,2010

272. PruchnoCJ, BurnsMM, SchulzeM, JohnsonRJ, Baker PJ, Alpers CE, Couser WG: Uri-nary excretion of the C5b-9 membrane at-tack complex of complement is a marker ofimmune disease activity in autologous im-mune complex nephritis. Am J Pathol 138:203–211, 1991

273. Schulze M, Pruchno CJ, Burns M, Baker PJ,Johnson RJ, Couser WG: Glomerular C3clocalization indicates ongoing immune de-posit formation and complement activa-tion in experimental glomerulonephritis.Am J Pathol 142: 179–187, 1993

274. Glassock RJ: The pathogenesis of idio-pathic membranous nephropathy: A 50-year odyssey.AmJKidneyDis 56: 157–167,2010

275. Beck LH Jr, Fervenza FC, BeckDM,BonegioRG, Malik FA, Erickson SB, Cosio FG,Cattran DC, Salant DJ: Rituximab-induceddepletion of anti-PLA2R autoantibodiespredicts response in membranous nephrop-athy. J Am Soc Nephrol 22: 1543–1550,2011

276. Penny MJ, Boyd RA, Hall BM: Role of T cellsin the mediation of Heymann nephritis. ii.Identification of Th1 and cytotoxic cellsin glomeruli. Kidney Int 51: 1059–1068,1997

277. Penny MJ, Boyd RA, Hall BM: PermanentCD8(+) T cell depletion prevents proteinuriain active Heymann nephritis. J Exp Med 188:1775–1784, 1998

278. Couser WG, Stilmant MM, Darby C: Au-tologous immune complex nephropathy. I.

Sequential study of immune complex de-position, ultrastructural changes, proteinuria,and alterations in glomerular sialoprotein.Lab Invest 34: 23–30, 1976

279. Stanescu HC, Arcos-Burgos M, Medlar A,Bockenhauer D, Kottgen A, DragomirescuL, Voinescu C, Patel N, Pearce K, HubankM,Stephens HA, Laundy V, Padmanabhan S,Zawadzka A, Hofstra JM, Coenen MJ, denHeijer M, Kiemeney LA, Bacq-Daian D,Stengel B, Powis SH, Brenchley P, FeehallyJ, Rees AJ, Debiec H, Wetzels JF, Ronco P,Mathieson PW, Kleta R: Risk HLA-DQA1and PLA(2)R1 alleles in idiopathic mem-branous nephropathy. N Engl J Med 364:616–626, 2011

280. Debiec H, Lefeu F, Kemper MJ, Niaudet P,Deschênes G, Remuzzi G, Ulinski T, RoncoP: Early-childhood membranous nephrop-athy due to cationic bovine serum albumin.N Engl J Med 364: 2101–2110, 2011

281. QinW, Beck LH Jr, ZengC, Chen Z, Li S, ZuoK, Salant DJ, Liu Z: Anti-phospholipase A2receptor antibody in membranous nephrop-athy. J Am Soc Nephrol 22: 1137–1143,2011

282. Abrera MA, Nishimura C, Frees K, Jones M,Maga T, Katz LM, Zhang Y, Smith RJ: Allelicvariants of complement genes associatedwith dense deposit disease. J Am SocNephrol 22: 1551–1559, 2011

283. Nasr SH, Valeri AM,AppelGB, Sherwinter J,StokesMB, Said SM,Markowitz GS, D’AgatiVD: Dense deposit disease: Clinicopatho-logic study of 32 pediatric and adult pa-tients. Clin J Am Soc Nephrol 4: 22–32,2009

284. Smith RJ, Harris CL, Pickering MC: Densedeposit disease. Mol Immunol 48: 1604–1610, 2011

285. Servais A, Frémeaux-Bacchi V, LequintrecM, Salomon R, Blouin J, Knebelmann B,Grünfeld JP, Lesavre P,Noël LH, Fakhouri F:Primary glomerulonephritis with isolatedC3 deposits: A new entity which sharescommon genetic risk factors with haemo-lytic uraemic syndrome. J Med Genet 44:193–199, 2007

286. Fakhouri F, Frémeaux-Bacchi V, Noël LH,Cook HT, Pickering MC: C3 glomerulo-pathy: A new classification. Nat Rev Nephrol6: 494–499, 2010

287. Falk RJ, Sisson SP, Dalmasso AP, Kim Y,Michael AF, Vernier RL: Ultrastructural lo-calization of the membrane attack complexof complement in human renal tissues. AmJ Kidney Dis 9: 121–128, 1987

288. Gale D, Pickering M: Regulating comple-ment in the kidney. Insights from CFHR5nephropathy. Dis Model Mech 4: 721–726,2011

289. Strobel S, Zimmering M, Papp K, Prechl J,Józsi M: Anti-factor B autoantibody indense deposit disease. Mol Immunol 47:1476–1483, 2010

290. Sethi S, Gamez JD, Vrana JA, Theis JD,Bergen HR 3rd, Zipfel PF, Dogan A, SmithRJ: Glomeruli of Dense Deposit Diseasecontain components of the alternative andterminal complement pathway. Kidney Int75: 952–960, 2009

291. Sethi S, Fervenza FC, Zhang Y, Nasr SH,Leung N, Vrana J, Cramer C, Nester CM,Smith RJ: Proliferative glomerulonephritissecondary to dysfunction of the alternativepathway of complement. Clin J Am SocNephrol 6: 1009–1017, 2011

292. Pickering MC, Cook HT, Warren J, BygraveAE,MossJ,WalportMJ,BottoM:UncontrolledC3 activation causes membranoproliferativeglomerulonephritis in mice deficient in com-plement factor H. Nat Genet 31: 424–428,2002

293. Abrera-Abeleda MA, Nishimura C, Frees K,Jones M, Maga T, Katz LM, Zhang Y, SmithRJ: Allelic variants of complement genesassociated with dense deposit disease.J Am Soc Nephrol 22: 1551–1559, 2011

294. Gale DP, de Jorge EG, Cook HT, Martinez-Barricarte R, Hadjisavvas A, McLean AG,Pusey CD, Pierides A, Kyriacou K, AthanasiouY, Voskarides K, Deltas C, Palmer A,Frémeaux-Bacchi V, de Cordoba SR, MaxwellPH, Pickering MC: Identification of a muta-tion in complement factor H-related protein5 in patients of Cypriot origin with glomeru-lonephritis. Lancet 376: 794–801, 2010

295. Rose KL, Paixao-Cavalcante D, Fish J,MandersonAP,Malik TH, BygraveAE, Lin T,Sacks SH, Walport MJ, Cook HT, Botto M,Pickering MC: Factor I is required for thedevelopment of membranoproliferative glo-merulonephritis in factor H-deficient mice.J Clin Invest 118: 608–618, 2008

J Am Soc Nephrol 23: ccc–ccc, 2012 Mediation of GN 19

www.jasn.org BRIEF REVIEW