You are on page 1of 13

REVIEWS

Genetics and pathogenesis of systemic lupus


erythematosus and lupus nephritis
Chandra Mohan and Chaim Putterman
Abstract | Systemic lupus erythematosus (SLE) is a multisystem autoimmune disorder that has a broad
spectrum of effects on the majority of organs, including the kidneys. Approximately 4070% of patients
with SLE will develop lupus nephritis. Renal assault during SLE is initiated by genes that breach immune
toleranceand promote autoantibody production. These genes might act in concert with other genetic
factorsthat augment innate immune signalling and IFN-I production, which in turn can generate an influx
of effector leucocytes, inflammatory mediators and autoantibodies into end organs, such as the kidneys.
The presence of cognate antigens in the glomerular matrix, together with intrinsic molecular abnormalities
in resident renal cells, might further accentuate disease progression. This Review discusses the genetic
insights and molecular mechanisms for key pathogenic contributors in SLE and lupus nephritis. We
have categorized the genes identified in human studies of SLE into one of four pathogenic events that
lead to lupus nephritis. We selected these categories on the basis of the cell types in which these
genes are expressed, and the emerging paradigms of SLE pathogenesis arising from murine models.
Deciphering themolecular basis of SLE and/or lupus nephritis in each patient will help physicians to tailor
specifictherapies.
Mohan, C. and Putterman, C. Nat. Rev. Nephrol. advance online publication 31 March 2015; doi:10.1038/nrneph.2015.33

Introduction

Department of
Bioengineering,
University of Houston,
3605 Cullen Boulevard,
Houston, TX 77204,
USA (C.M.). Division of
Rheumatology, Albert
Einstein College of
Medicine, 1300 Morris
Park Avenue, Bronx,
NY10461, USA (C.P.).
Correspondence to:
C.M.
cmohan@
central.uh.edu

Systemic lupus erythematosus (SLE) is a chronic auto


immune inflammatory disease that can affect the major
ity of organs and tissues. The clinical presentations
ofSLE can range from mild to severe and the course of
the disease is unpredictable, with periods of remission
and flares. Lupus nephritis is a severe consequence of
SLE and an important driver of morbidity and mortality
inSLE. Lupus nephritis affects ~4070% of patients with
SLE, with the exact incidence dependent on ethnicity, age
group, and gender. Both systemic and intra-renal events
are important in the pathogenesis of lupus nephritis.14
At the systemic level, both the adaptive and innate
branches of the immune system contribute to the devel
opment of SLE. The two predominant cell types involved
in the adaptive immune system, B lymphocytes (Bcells)
and T lymphocytes (Tcells), are both essential for the
development of lupus nephritis.26 Bcells are pathogenic
in SLE because of the autoantibodies (for example, antiDNA antibodies and anti-nucleosome antibodies) and
cytokines that they produce.7,8 Tcells drive the systemic
and intra-renal activation of Bcells. Subtypes of Tcells,
including type1 Thelper (TH1) cells, type17 T-helper
(T H17) cells, and CD3 +CD4 CD8 double negative
THcells, have been implicated in the pathogenesis of
lupus nephritis.911 The innate immune system contrib
utes to the pathogenesis of SLE in multiple ways. In the
early stages of disease, dendritic and other myeloid cells
Competing interests
The authors declare no competing interests.

activate Tcells, and produce key mediators such as B-cell


activating factor; this effect results in the activation of
the adaptive immune system.1214 Activation ofthe sys
temic immune system leads to the generation of effec
tor Tcells and autoantibodies that can subsequently
target organs, such as the kidneys.13,15,16 These effectors,
together with numerous soluble mediators, elicit chronic
inflammation within glomerular and tubulointerstitial
sites in the kidneys. By contrast, the contribution of
intrinsic renal cells versus systemic leucocytes in the
pathogenesis of lupus nephritis is poorly understood.
Detailed reports describing the systemic and intra-renal
events in the pathogenesis of lupus nephritis have been
reviewedelsewhere.6,13,1520
The present Review categorizes the genes implicated
in SLE according to the cell types and molecular path
ways in which they are expressed, to gain insight into the
key pathogenic contributors that lead to lupus nephritis.
We first introduce the methods by which novel genetic
loci have been identified and the functional studies
performed to characterize candidate genes. Next, we
discuss the genes that are implicated in the activation
of the adaptive and innate immune systems, as well as
those that are involved in the intra-renal processes that
promote tissue damage. We conclude by discussing the
potential mole cules that might affect the amount of
accessible chromatin that is generated from apoptotic
cells. While the organization of SLE and lupus nephri
tis candidate genes into these particular pathways is not
fixed and alternate classification schemes might also be

NATURE REVIEWS | NEPHROLOGY

ADVANCE ONLINE PUBLICATION | 1


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Key points
Some genes implicated in systemic lupus erythematosus (SLE) and lupus
nephritis might contribute to the pathology of disease by breaching immune
tolerance and promoting autoantibody production
A subset of SLE and/or lupus nephritis genes might augment innate immune
signalling and IFN-I production; other SLE genes might modulate the molecular
pathways that lead to renal tissue damage
Genes that affect the accessibility and handling of apoptotic material and
chromatin might also contribute to SLE and/or lupus nephritis
The presence of cognate antigens on the glomerular matrix, together with
intrinsic molecular abnormalities in resident renal cells, could further
accentuate disease progression in lupus nephritis
Differential involvement of the above-listed mechanisms in patients could
potentially explain the wide spectrum of clinical phenotypes observed among
individuals with SLE and lupus nephritis

suitable, we find the following categories to be a useful


framework for understanding the genetic contributions
to SLE and lupus nephritis.

Gene discovery
Genome-wide association studies
Genome-wide association studies (GWAS) that aimed to
identify genetic loci linked with SLE were initiated more
than 6years ago.1721 Thus far, >10GWAS have been con
ducted using samples obtained from patients with SLE
that encompass multiple ethnic groups, and have collec
tively identified >50genes associated with SLE. Although
the focus of these GWAS has not been exclusive to lupus
nephritis, the inclusion of patients with lupus nephritis
in these studies has provided important insights into the
potential pathogenic pathways that lead to both SLE and
lupus nephritis. Several genes identified from GWAS
andadditional candidate genes have been validated in
independent patient cohorts as being associated with SLE
or lupus nephritis. Table1 lists some of the key genes that
have been implicated in the pathogenesis of SLE and/or
lupus nephritis; this list is not exhaustive and additional
studies are ongoing.
Genetic aberrations implicated in SLE
Although numerous genes have been implicated in SLE
and/or lupus nephritis (Table1), questions remain as
to their function in disease pathology. Firstly, the spe
cific causative mutations and subsequent molecular
alterations that contribute to the disease phenotype
have not been firmly established for many of the identi
fied candidate genes. Coding region single nucleotide
polymorphisms (SNPs), polymorphisms that lead to
differential alternative splicing, polymorphisms in the
3'untranslated region (UTR) that influence gene expres
sion, and copy number variants (CNVs) have all been
documented. This catalogue of implicated molecular
variations in each gene is likely to expand as more infor
mation becomes available from deep sequencing studies
that are currently in progress. Secondly, the association
between these candidate genes and SLE or lupus nephri
tis has only been inferred from studies using murine
models that have been engineered to either lack or
overexpress the gene (using knockouts or transgenics,

respectively); some of these associations have been


reviewed elsewhere.22 Additional murine models should
be generated that mimic more closely the type of genetic
mutations that are observed among human patients with
SLE and lupus nephritis; these specific mutations (SNPs,
CNVs and splice isoforms) should be subsequently
validated for their ability to promote SLE in genetically
engineered murine models.
Categorizing genes implicated in SLE
The identified genes implicated in SLE can be assigned to
one of four functional categories: genes that affect lym
phocyte activation, particularly Bcells; genes that affect
innate immune signalling, notably NF-B activation and
IFNI signalling; genes that might function within the
kidneys, potentially promoting renal tissue damage; and
genes that influence the handling of apoptotic debris,
chromatin, and immune complexes bearing these anti
gens. These categories have been designated on the basis
of apriori information regarding the cell types in which
the identified genes are expressed and their known
molecular function; however, alternative pathways and
models cannot be excluded.
Genetic composition of SLE and lupus nephritis
Although a given patient might harbour mutations in
multiple genes from a given functional category, whether
the inheritance of one or more genes from each category
proposed above is required for the development of SLE
and/or lupus nephritis is unclear. Murine modelsof
SLE and/or lupus nephritis have been insightful in
this respect as genes from all four categories have been
implicated through both forward and reverse genetic
approaches.2326 Congenic dissection (where multiple
genes mediating a polygenic disease are separated into a
collection of congenic strains) and congenic reconstitu
tion studies (involving genetic crossing of each separate
congenic strain) of SLE-associated polymorphic vari
ants have generated three main findings. Firstly, genes
that only affect lymphocyte function might lead to the
production of antinuclear antibodies or IgM polyreac
tivity but not lupus nephritis, suggesting that simply the
presence of these antibodies is not sufficient for renal
pathology to ensue. Secondly, genes that activate innate
immune signalling in isolation might result in the pro
duction of intermediate levels of anti-DNA antibodies
(compared to polygenic strains with complete SLE) and
non-proliferative glomerulonephritis. Finally, the epi
static interaction between genes from multiple categories
is required for severe lupus nephritis to develop.2730 The
emerging concept of genetic interaction is exemplified by
a series of congenic dissection and reconstitution studies
that have been performed using the C57Bl/6 (B6) mouse
strain, as illustrated inFigure1.27,2932
Through extrapolation of the genetic data obtained
from murine congenic dissection and reconstitution
studies, it could be predicted that genes from mul
tiple functional categories would act in concert to
promote lupus nephritis in human patients. Although
it is apparent which functional pathways the majority of

2 | ADVANCE ONLINE PUBLICATION

www.nature.com/nrneph
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Table 1 | Susceptibility genes implicated in human SLE or LN*
Gene name

Association with SLE

Association with LN

BANK1

Yes168170

Unknown

BLK

Yes169175

Yes70

CD80

Yes63

Unknown

CSK

Yes

Unknown

ETS1

Yes169,172,174,175

Unknown

HLA DR

Yes

Yes70,71

IL-10

Yes

Unknown

LYN

Yes178

Unknown

PP2A

74

Yes

Unknown

PRDM1 (BLIMP1)

Yes177,179

Unknown

PRKCB

Yes180

Unknown

PTPN22

Yes

Unknown

RasGRP3

Yes172,184

Unknown

STAT4

Yes170,172,174,175,185,186

Yes70,72

TNFSF4 (OX40L)

Yes

Yes76,77

IFIH1

Yes188,189

Unknown

IKZF1

Yes172,175,189,190

Yes81

ILT3

Yes24

Unknown

IRAK1

Yes

Unknown

IRF5

Yes23,169,172,175,193196

Yes70,83

IRF7

Yes197,198

Unknown

IRF8

Yes

Unknown

TLR7

Yes201,202

Unknown

TLR9

Yes203

Yes87

TNFAIP3 (A20)

Yes

Yes101

TNIP3 (ABIN3)

Yes172,177

Yes94

TYK2

161,189

Yes

Unknown

UBE2L3

Yes205,206

Unknown

ACE

Yes125

Yes125

COL25A1

Yes

Unknown

KLK

Yes127

Yes127

LAMC2

126

Yes

Unknown

ATG5

Yes179

Unknown

DNAse1

Yes

Unknown

FCGR2A, 3A, 3B

Yes95100,170,186

Yes96

ITGAM

Yes

Yes77,102,106

TREX1

Yes

Unknown

Lymphocyte signalling

50

58,59,174
176,177

51,53,181183

169,170,172,175,187

Innate immune signalling

107,191,192

189,199,200

21,169,172,174,175,204

Intra-renal signalling

126

Immune complex clearance

207

170,186,208210
211

*Susceptibility genes implicated in SLE or LN have been functionally organized into four pathogenic
cascades. Additional genes associated with SLE have not been listed but are reviewed elsewhere. 25
Abbreviations: LN, lupus nephritis; SLE, systemic lupus erythematosus.

SLE-associated genes are most likely to impact, for some


genes the functional pathway is unclear. For example,
complement genes and proteins could potentially
exert effects on several pathways, including chromatin
handling and B-cell activation.

Adaptive immune system activation


Modulation of Bcell receptor signalling
Tcells, Bcells and autoantibodies have been shown to
have an essential function in lupus nephritis, with the
majority of the evidence originating from mechanistic
studies performed in mice.1,1720 A conglomerate of SLEassociated genes collectively regulates B-cell signalling
via the Bcell receptor, BCR (Figure2). BANK1 is a
scaffold adaptor protein primarily expressed in Bcells
that amplifies Bcell signalling by linking the activa
tionof Src-family kinases, such as LYN, with the acti
vation ofthe IP3 receptor, leading to calcium influx.
Stimulationof BCR facilitates the association of BANK1
with PLC2, resulting in PLC2 activation, expression
of IP3 and DAG, and subsequent activation of PRKCB,
another gene implicated in SLE.3336
RasGRP3one of the downstream molecules acti
vated by the BCR pathwayhas also been implicated
in SLE (Figure2). RasGRP3 amplifies downstream Rasmediated ERK and MAPK signalling in Bcells, and is
associated with B-cell proliferation and antibody pro
duction. PRKCB and RasGRP3 are expressed in Bcells,
Tcells, myeloid cells, and endothelial cells, and thus
could contribute to the progression of SLE through
several different pathways.3741 Although genetic manip
ulation studies of Bank1 and Rasgrp3 are yet to be pub
lished, the available data suggest that PRKCB might be
necessary for the development of SLE and lupus nephritis
in mice.42
Src-family tyrosine kinases
Three SLE-associated B-cell genes, LYN, BLK and CSK,
are Src-family tyrosine kinases that might have direct or
indirect inhibitory roles on BCR signalling.4246 The fact
that Lyn-deficient mice develop B-cell hyperactivity with
associated autoimmunity is well documented.4446 The
inhibitory effect of LYN on B-cell signalling is mediated
in part by phosphorylation and subsequent activation
of the inhibitory B-cell surface receptors, CD22 and
FcGR2B. The CD22 and FcGR2B receptors function
through SHIP and SHP-1 phosphatases and dampen
B-cell activation as a consequence.43
One function of BLK is to promote the interaction
between BANK1 and PLC2 (Figure 2). Reduced
expression of BLK is associated with reduced numbers
of pre-Bcells and marginal zone Bcells. Reduced expres
sion of BLK can also promote renal disease in B6 mice
that are prone to SLE.47,48
The third Src-family kinase, CSK, can phosphory
late and regulate LYN. 49 Overexpression of CSK is
observed among patients with SLE that carry the CSK
risk allele, and leads to increased phosphorylation of
LYN; this effect is associated with elevated production
of antibodies and transitional Bcells.50
PTPN22
PTPN22 has been implicated in several autoimmune
diseases, including SLE, and the effect of a disease-
associated variant of Ptpn22 on systemic autoimmun
ity has been verified using murine models.51 PTPN22

NATURE REVIEWS | NEPHROLOGY

ADVANCE ONLINE PUBLICATION | 3


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Wild-type
C57Bl/6

a
Gene 1
e.g. Ly108

Adaptive
immunity

b
Gene 2
e.g. Sle3

Ly108

Innate
immunity
Ly108
Sle3

c
Gene 3
e.g. Klk

End-organ
disease
Ly108
Sle3

Klk

Figure 1 | Stepwise evolution of systemic lupus erythematosus (SLE) as a function of genetic load. The schematic depicts
Nature Reviews | Nephrology
how genetic dissection studies in the mouse have been used to investigate the evolution and pathology of SLE. 27,2931
Thegenetic intervals Sle1 and Sle3 were initially mapped in the NZM2410 lupus-prone strain, and subsequently bred to the
C57Bl/6 (B6) background as congenic intervals. These genetic dissection studies demonstrated that genes associated
with SLE might contribute to disease pathogenesis in multiple ways, including breaching lymphocyte tolerance, activating
innate immunity and regulating inflammation within the end organs. a | Ly108, identified as a culprit gene within the Sle1
genetic interval, was introgressed onto the C57Bl/6 background, resulting in activation of the adaptive immune system.
b | The addition of the Sle3 genetic interval, which activates innate immunity, results in robust autoimmune phenotypes in
the bicongenic strain. c | Finally, the presence of genes (for example, Klk) that regulate function in the end-organs, such
asthe kidneys, leads to fully developed SLE.

regulates CSK and can modulate the BCR and T-cell


receptor signalling threshold and downstream activa
tion of AKT in both Bcells and Tcells.3236,42 In addition
to activating Bcells, PTPN22 also modulates the extent
of the regulatory T (TREG) cell pool, and affects myeloid
cell-signalling.5255 Specifically, the numbers of thymic
and TREG cells vary inversely with the levels of PTPN22,54
whereas PTPN22 deficiency reduces IFN-I production
and augments inflammation.55
At present, it is not fully understood how LYN, BLK,
CSK and PTPN22 interact to fine-tune BCR signalling, or
how SNPs in various genes within this axis might impact
the overall strength of the signalling cascade. Given that
SNPs in all of the above genes can modulate the extent
of BCR signalling, it would be interesting to determine
whether an increasing number of genetic hits in this
pathway confer increased risk for SLE or lupus nephritis;
data to support this concept are currentlylacking.
Interaction of BCRs with Toll-like receptors
Interaction between the signalling pathways triggered
by the BCR and Toll-like receptors (TLRs) can serve
to amplify B-cell signalling. SLE autoantigens, such as
DNA and RNA, can also bind to TLRs such as TLR9
and TLR7.56 This interaction in turn activates a signal
ling pathway that is shared with innate immune cells,
involving MYD88, IRAK4 and IRAK1, which eventu
ally leads to the activation of NF-B, IRF5, and IRF7
(Figure2). These transcription factors are responsible for
the increased survival of Bcells, autoantibody produc
tion and the production of several cytokines, including
IFN-I. Importantly, several molecules in this pathway
constitute SLE-associated genes, including TLR7, IRAK1,
IRF5, IRF7, and molecules that regulate NF-B activa
tion such as UBE2L3, TNFAIP3 and TNIP3 (Figure2).
These genes will be discussed in the analysis of the innate
immunesystem.

Cross-talk between Tcells and Bcells


SLE-associated genes might mediate cross-talk between
lymphocytes. Bcells are able to endocytose and process

the autoantigens that they bind to, and subsequently


present autoantigen-derived peptides to T H cells
through their MHC-II molecules, as demonstrated for
nucleosome-derived histone epitopes.57 MHC-II mol
ecules, notably HLA-DR2 and HLA-DR3, represent
two of the best characterized genes with the strongest
association with SLE;58,59 however, the available infor
mation with regard to their role in SLE is difficult to
interpret. Multiple HLA-DR2 and HLA-DR3 alleles, as
well as several additional HLA alleles, are associated
with SLE. The strength of the association and specific
allele(s) implicated in SLE depends on the ethnic group
and clinical presentation being studied.5860 Different
SLE-associated HLA MHC-II alleles have documented
associations with other autoimmune diseases, such as
diabetes mellitus and Sjgren syndrome.60
Co-stimulatory molecular pairs, such as CD28
CD80, CD40CD40L and OX40OX40L (also known as
TNFSF4), also have an important function in the bilat
eral amplification of lymphocyte cross-talk (Figure2).61
Among these co-stimulatory molecules, OX40L and
CD80 have been implicated in susceptibility to SLE
(Table1).1720,62,63 Interaction between OX40 and CD123
or TNFSF4 on activated Tcells might result in the gener
ation of IL-10-producing TREGcells.62 These same pairs of
co-stimulatory molecules might also regulate the inter
action between specialized antigen-presenting cells, such
as dendritic cells, and Tcells (Figure2).

Generation of immune effector cells


The activation of Bcells and Tcells leads to the gen
eration of effector cells, which can subsequently cause
tissue injury. Such effector cells include plasma cells and
TH17cells, both of which have been implicated in the
pathogenesis of lupus nephritis.6467 ETS1 and PRDM1
(also known as BLIMP1) are two molecules that can regu
late the generation of both plasma cells and TH17cells.
Evidence suggests that expression of the mRNA transcript
for the negative regulator ETS1 is reduced in peripheral
blood mononuclear cells of patients with SLE, and Ets1
ablation leads to SLE in mice.68,69 Conversely, blockade

4 | ADVANCE ONLINE PUBLICATION

www.nature.com/nrneph
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Nuclear antigen
B-cell receptor

OX40L*

OX40

BANK1*
BLK1*
PLC2

IP3
CD40
MHCII*

DAG

CD40L

Myd88
TRAF6
IRAK4
IRAK1*

PRKCB*

TCR
PTPN22*

RasGRP3*

CSK*
Antigenpresenting
cell
(dendritic
cell)

Endosome
TLR8

CD28

B cell

PTPN22*

TLR9*

CD80*

CSK*
LYN*

B-cell
receptor
signalling

TLR7*

T cell

Ras

STAT4*

ERK

PP2A*

UBE2L3*
TNFAIP3*

NF-B

IRF5*

IRF7*

TNIP3*
Nucleus

ETS1*
PRDM1*
TH17
cells

Increased survival
antibody production

Plasma
cells

Cytokine
production

IFN-I
production

Figure 2 | Gene products associated with systemic lupus erythematosus (SLE) that might affect the
adaptive
immune
system.
Nature
Reviews
| Nephrology
The schematic shows some of the interactions and signalling pathways involving known SLE genes (denoted by an asterisk)
that have the potential to influence lymphocyte function. These genes have been divided into three groups, depending on the
cell type in which they are expressed: antigen-presenting cells (dendritic cells), Tcells, and Bcells. The overall result of
signalling pathways in these three cell types is the production of effector cells, including TH17cells and plasma cells. The
black arrows represent activation and bar-headed lines represent inhibition of the target gene or process. The dashed lines
represent the overall response. Abbreviations: TCR, T-cell receptor; TH17 cells, Thelper 17 cells; TLR, Toll-like receptor.

or deficiency of PRDM1 reduces the severity of SLE in


mice.70,71 Both genetic evidence from human GWAS and
mechanistic studies in mice, therefore, underscore the
importance of the generation of TH17cells and plasma
cells in the pathogenesis of SLE.
Both mouse models of SLE and patients with SLE
exhibit increased levels of IL-17 production and increased
numbers of circulating TH17 cells that correlate with
disease activity.72 Furthermore, IL-17 deficiency pro
tects mice from the development of SLE-associated glo
merulonephritis, although conflicting evidence has also
been reported.72,73 Polymorphisms in PPP2A detected in
patients with SLE are associated with elevated expres
sion of the PP2A catalytic subunit (PP2Ac) in Tcells and
decreased IL-2 production, which can be regulated by
miRNA155.74,75 A transgenic mouse overexpressing Pp2ac
showed increased T-cell production of IL17 and high
susceptibility to the development of antibody-mediated
nephritis. Blockade of IL17by intraperitoneal antibody
administration markedly reduced proteinuria and histo
logical damage in this transgenic mouse model.76 PP2Ac
has been shown to deregulate the IL-17 locus via enhanced
histone 3 acetylation; this effect was identified in both
Tcells of Pp2ac transgenic mice and in patients with SLE.77
Some of the disease genes in this category have already
shown an association with lupus nephritis, as summa
rized in Table1. These associations include SNPs in BLK,

HLA DRB1, STAT4 and OX40L. The presence of SNPs in


OX40L and expression levels of OX40L on the surface of
peripheral blood mononuclear cells are both associated
with lupus nephritis.62,7884

Activation of innate immune signalling


Modulation of IFNI signalling
SLE in both paediatric and adult patients is increas
ingly recognized to have a molecular signature that is
characterized by over-activation of the IFN-I signal
ling pathway.85 IFN-I signalling is important in myeloid
cells, including monocytes and dendritic cells, and might
also have important functions in resident renal cells.86,87
Previous studies suggest a possible genetic basis for the
increased expression of IFN-I observed in SLE; SNPs in
several genes in this pathway are associated with SLE
(Table1). Signalling via the IFN-I receptor is regulated
by JAK1, TYK2 and various STAT proteins, includ
ing STAT4. Importantly, SNPs in TYK2 and STAT4 are
associated with SLE (Figure3). Another SLE-associated
gene, IKZF1, regulates the transcription of STAT4, and
polymorphisms in IKZF1 are also associated with lupus
nephritis (Figure3).88
JAKSTAT signalling
Activation of STAT4 occurs not only in lympho
cytes but also in activated monocytes and dendritic

NATURE REVIEWS | NEPHROLOGY

ADVANCE ONLINE PUBLICATION | 5


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Immune
complex

FCGR2A*

IL-12
IL-23
IFN-I

FCGR3A*
FCGR3B*

ITGAM*

IFNAR
JAK1
Endosome

LYN*

TLR9*

TLR7*

Multiple
signalling
pathways

TLR8

IKZF1*
Activation
and
phagocytosis

TYK2*

STATs
STAT4*
IFN-I-induced
genes
IRF5*
IRF7*
IFIH1*
ILT3*

Myd88
TRAF6
IRAK4
IRAK1*
UBE2L3*
TNFAIP3*

NF-B

IRF5*

IRF7*

TNIP3*
Nucleus
Myeloid cell

Increased
survival

Cytokine
production

IFN-I
production

Nature
Reviews | Nephrology
Figure 3 | Gene products associated with systemic lupus
erythematosus
(SLE)
that might affect innate immune signalling. The schematic illustrates the
signalling pathways that occur in myeloid cells that involve genes that could
influence innate immune cell function (denoted by an asterisk). Three myeloid cell
surface receptors (ITGAM, FcR family receptors and IFNAR) are illustrated from
which signalling cascades are initiated upon presentation of immune complexes.
The overall response is regulation of gene transcription that results in the
modulation of cell survival and the production of cytokines and IFNI. The black
arrows represent activation and bar-headed lines represent inhibition of the target
gene or process. The dashed lines represent the overall response. Abbreviation:
TLR, Toll-likereceptor.

cells; indeed, monocytes from the kidneys of patients


with autoimmune diseases also express high levels of
STAT4.8990 Although the JAKSTAT pathway has been
shown to mediate the progression of renal fibrosis, the
specific disease contributions of TYK2, STAT4 and
IKZF1 in intrinsic renal cells are currently unknown.
Interestingly, patients with SLE who harbour SNPs in
STAT4 develop symptoms of the disease at an earlier
age compared to patients who do not carry these
SNPs; these patients also exhibit high levels of antidsDNA antib odies and develop severe lupus nephri
tis.80 Furthermore, Stat4 deficiency in mice aggravates
lupus nephritis, as compared to mice that are Stat4sufficient.81 Given that diverse cell types express STAT4,
dissecting the cellular and molecular mechanisms by
which SNPs in this gene contribute to lupus nephritis
poses a considerablechallenge.
Activation of IFN receptor by interleukins
IL-12 and IL-23 can activate the IFN / receptor
(IFNAR), which results in increased production of
IFN-I.91,92 Consequently, a large number of genes that
are regulated by IFN-I become activated, constituting

the most prominent SLE-associated RNA signature.71


Some of these IFN-I-regulated genes, such as IRF5, IRF7,
ILT3 and IFIH1, are SLE-associated genes, andsome
can in turn regulate the expression of IFN-I and other
disease-associated cytokines.93 Of note, the IRF5 allele
implicated in SLE is associated with high serum IFN-I
activity and the development of anti-dsDNA anti
bodies.23 Similarly,the ILT3 disease-associated allele
correlateswith elevated IFN-I and TNF expression
among patients with SLE.24 The increased production of
IFN-I that is driven by these disease alleles is expected
to initiate a positive feedback loop that rapidly increases
IFN-I production (Figure3). The pathogenic importance
of this axis is also underscored by the observation that
genetic ablation or pharmacological blockade of key
molecules in this axis, such as IRF5, suppresses SLE in
mice.25 Furthermore, SNPs in IRF5 are associated with
lupus nephritis in patients.26,70 Finally, emerging evi
dence suggests that genetic aberrations in multiple genes
within the IFNI pathway might confer increased disease
susceptibility, as exemplified by the genetic interaction
between IRF5, IKZF1 andSTAT4.94

Fc receptors for immunoglobulin


The Fc receptors (FcRs) for IgG represent another
family of receptors that have a prominent role in the
activityof myeloid cells. SNPs and CNVs affecting
multiple members of this family, including FCGR2A,
FCGR3A and FCGR3B, have been associated with SLE
and LN.95100 Interestingly, some of the SNPs and CNVs
in FcRs associated with lupus nephritis are characterized
by reduced FcR expression on the myeloid cell surface
membrane; this observation has led to the hypothesis
that reduced FcR-mediated immune complex clearance
might contribute to lupus nephritis.9698 Total ablation
of FcRs, however, prevents the development of lupus
nephritis despite the presence of anti-DNA antibodies.101
These data suggest that myeloid cells are the major cell
type through which FcR expression can influence the
pathogenesis of lupusnephritis.102
FcRs not only activate myeloid cells, but also enable
these cells to endocytose immune complexes.103 Immune
complexes present in SLE might harbour nuclear anti
gens, such as RNA and DNA, allowing the endocytosed
immune complexes to activate endosomal TLRs such
as TLR7, TLR8 and TLR9.104 Activation of these TLRs
recruit MYD88 and a series of additional molecules,
including IRAK4, IRAK1 and TRAF6, which together
initiate various transcriptional programmes, including
those driven by NF-B, IRF5 and IRF7 (Figure3).105
These transcriptional processes have important func
tions in augmenting proinflammatory cytokine pro
duction, including IFN-I. Several of these molecules,
including TLR7, TLR9, IRAK1, IRF5 and IRF7 have
been implicated as SLE-causative genes, among which,
TLR9 and IRF5 have been directly associated with lupus
nephritis in patients.26,106 In addition, genetic ablation or
pharmacological blockade in mouse models of SLE have
established a pathogenic role for Tlr7, Irak1 and Irf5 in
this disease.25,107109 Enhancing Tlr7 expression through

6 | ADVANCE ONLINE PUBLICATION

www.nature.com/nrneph
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
Immune
complex

FCGR3A*

FCGR3B*

FCGR2A*

FCGR2A*
FCGR3A*

Glomerular
matrix

FCGR3B*

TREX1*

Endosome

Myd88
TRAF6
IRAK4
IRAK1*

ITGAM*

UBE2L3*
TNFAIP3*

KLK*

NF-B

IRF5*

IRF7*

TNIP3*

ACE*

COL25A1*

Intra-renal promotion of tissue damage

Nucleus

LAMC2*
Myeloid
cell

TLR7*

Multiple
signalling
pathways

C1*
C2*
C3*
C4*

TLR8

Fc

TLR9*

DNAse1*

have also shown associations with lupus nephritis in


humanpatients.114,115
ITGAM (also known CD11B or MAC1) is a myeloid-
specific adhesion molecule with multiple ligands, includ
ing ICAM1, ICAM2, C3bi and FGA. 116118 ITGAM
exhibits diverse functions under normal physiological
conditions, including mediating myeloid-cell activation
(leading to production of NF-B, IL-6 and TNF), phago
cytosis, uptake of immune complexes, and facilitating
interactions between myeloid cells, lymphocytes, plate
lets and endothelial cells.116118 The ITGAM Arg77His
allele has reduced binding efficiency for ICAM1, and
thus has a potential effect on cell adhesion. The Arg77His
allele correlates with severe disease manifestations of
SLE, including lupus nephritis, as well as h
aematological
and neurological phenotypes.84,119,120

Resident
renal cell

Increased
survival

Cytokine
production

IFN-I
production

Figure 4 | Gene products associated with systemic lupus


erythematosus
(SLE)
Nature
Reviews | Nephrology
that might affect intra-renal events leading to lupus nephritis. The schematic
depicts genes associated with SLE that might exert effects on intra-renal events
that lead to lupus nephritis (denoted by an asterisk). Myeloid cells present to
resident renal cells via Fc receptors, which act in concert with complement
molecules (C1C4). Upon stimulation by an immune complex, resident renal cells
can initiate multiple signalling pathways that culminate in the activation of NF-B,
IRF5 and IRF7. In this way, gene transcription can be modulated, with subsequent
effects on cell survival, cytokine production and expression of IFN-I. The black
arrows represent activation and bar-headed lines represent inhibition of the target
gene or process. The dashed lines represent the overall response. Abbreviation:
TLR, Toll-like receptor.

gene duplication confers susceptibility to SLE and lupus


nephritis in mice.108,109 Multiple lines of evidence there
fore support the critical importance of this molecular
axis in the pathogenesis of SLE and lupusnephritis.

Modulation of NF-B activation


Activation of NF-B has been implicated in both SLE and
lupus nephritis. NF-B can be negatively regulated by
TNFAIP3 (also known as A20) and TNIP3 (also known
as ABIN3), either independently, or in concert.110 TNIP3
can bind to TNFAIP3 to facilitate NFB inhibition.110
UBE2L3 is an E2 ubiquitin-conjugating enzyme that
can regulate the level of NFB activation and result
ant cell proliferation.111 SNPs in TNFAIP3, TNIP3 and
UBE3L3 have shown an association with SLE in human
patients (Figure3).112 Whether multiple perturbations
to this pathway have an incremental effect on NFB
activation remains to be explored. Murine studies have
offered further evidence to support the importance
of this pathway in SLE progression. Reduced expres
sion of TNFAIP3 in Bcells augments the production of
autoantibodies and germinal centre type Bcells, and the
development of renal disease.113 TNFAIP3 and TNIP3

Activation of Tcells and Bcells in the kidney


Genes associated with SLE can contribute to the patho
genesis of lupus nephritis either directly or indirectly.
Cells involved in the adaptive arm of the immune system
(Bcells and Tcells) are known to infiltrate the kidneys.
Previous work has documented the existence of struc
tures reminiscent of germinal centres and other lym
phocyte clusters within the inflamed kidneys of patients
with lupus nephritis.65 All of the genes described above
in the adaptive immune system can potentially influence
lupus nephritis by activating Tcells and Bcells, both
systemically and within the kidneys.
Activation of intra-renal myeloid cells
Cells of the innate immune system, such as macrophages
and neutrophils, are present in large numbers within
the kidneys of patients with lupus nephritis. All of the
genes known to affect innate immune signalling, which
were outlined in the section describing the innate
immune system, can potentially influence the progres
sion of lupus nephritis by activating intra-renal myeloid
cells.121,122 In addition, some of these genes, notably
ITGAM and FcR, could potentially affect the level of
recruitment of myeloid cells to the glomerular matrix
via cognate binding sites on the immune complexes
deposited in the glomeruli.122 Polymorphic variants of
ITGAM and/or FcR that result in increased binding to
complement components (for example, C3b) or the Fc
fragment of IgG could possibly augment myeloid cell
recruitment and activation (Figure4); however, the
biochemical and mechanistic evidence to support this
hypothesis is currently lacking. Conversely, evidence
exists to support reduced levels of FcR and reduced FcR
binding ability to IgGs among patients with SLE, as will
be discussedbelow.
Immune complex clearance from the glomeruli
Evidence suggests that SLE-associated genetic poly
morphisms might regulate the clearance of immune
complexes from glomerular tissue. SLE-associated SNPs
in FCGR2A (Arg131) and FCGR3A (Phe158) might
impair affinity for IgG binding.95,96 Likewise, CNVs in

NATURE REVIEWS | NEPHROLOGY

ADVANCE ONLINE PUBLICATION | 7


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
FCGR3B could reduce the cell surface expression levels
of this receptor.98100 Both of these effects could act to
reduce the uptake and clearance of immune complexes
from the glomerular matrix, not only by the infiltrating
myeloid cells, but also by resident mesangial cells, which
are known to upregulate the same FcRs upon stimula
tion with IFN-. The effect of FcR expression on infil
trating myeloid cells seemed to be more important in
the pathogenesis of lupus nephritis as compared to the
effect of expression of FcR on r enal-resident cells, in a
direct comparison.102

Glomerular deposition of nuclear material


Genes associated with SLE might augment the amount
and accessibility of nucleosomal material available for
deposition on the glomerular matrix, as will be discussed
further in the next section. Strong experimental evidence
supports the notion that nuclear antigens expressed
within the glomerular matrix can serve as points of inter
action with anti-nuclear antibodies (Figure4).123 Genes
that regulate DNA turnover (for example, DNASE1 and
TREX1), autophagy, and complement levels, therefore,
might regulate the amount of nuclear material that
is deposited on the glomerular matrix, as well as the
subsequent accumulation of immune complexes and
leucocyterecruitment.124
Genetic regulation of resident renal cells
SLE-associated genetic polymorphisms might regu
late specific disease pathways within resident renal
cells, including mesangial cells, podocytes, glomerular
endothelial cells and tubular epithelial cells. Potential
SLE genes in this category include ACE,125 extracellular
matrix molecules (COL25A1 and LAMC2),126 KLK127
and genes that activate the IFN-I, TLR and NF-B
signalling pathways (Figure4). Kidneys damaged by
nephritis express multiple TLRs; 128,129 furthermore,
renal cells grown in culture, such as mesangial cells,
podocytes, glomerular endothelial cells and tubular epi
thelial cells, express multiple TLRs and are responsive
to cognate TLR ligands.129135 Expression of the innate
signalling axes downstream of TLR activation, including
MYD88, TRAF6 and TNFAIP3, has been documented in
renalcells.136139
SLE genes that influence immune signalling could
impact disease in two ways: firstly, by affecting the
infiltration of leucocytes, and secondly, by affecting
the intrinsic properties of renal cells. Another possi
bility is that some SLE genes might produce differen
tial effects systemically and intra-renally, as illustrated
by CD80. CD80 usually mediates leucocyte cross-talk
in the immune system but has a dramatically different
function within podocytes.140142 Specifically, stimula
tion of TLR3 and TLR4 results in upregulation of CD80
in podocytes in an NF-B-dependent manner, which
leads to abrogation of the glomerular filtration barrier
and consequent proteinuria.141 Nephropathies charac
terized by this molecular feature could be treated with
selected biologic therapies,143 although the underlying
mechanisms require further exploration.

Novel lupus nephritis candidate genes


In addition to the genes discussed above, further mol
ecules might be important in mediating lupus nephri
tis that have not yet been implicated as disease genes in
GWAS. One such example is CAMK4. In mice with SLE,
inhibition of CAMK4 is associated with decreased IL2
production in Tcells, decreased cell proliferation, and
decreased IL6 production in PDGF-stimulated mesan
gial cells.144 Ablation of Camk4 in the lupus-prone murine
strain MRL/lpr, resulted in reduced glomerulonephritis,
accompanied by reduced cytokine production by Tcells
and macrophages.144 Other murine studies have also been
informative in this respect. A series of congenic dissection
studies in the New Zealand mixed strain SLE mouse model
(NZM2328) have facilitated the separation of genes that
contribute to autoantibody production from those that
promote acute glomerulonephritis, chronic glomerulo
nephritis and end-stage renal disease.145147 A 1.34Mb
region containing 45 genes at the distal end of chromo
some 1 (the Cgnz1 locus) was associated with chronic glo
merulonephritis, whereas the corresponding allele from
non-autoimmune mice conferred resistance to end-organ
damage. Although the exact gene within the Cgnz1 locus
is yet to be identified, this genomic region contains several
potentially interesting candidate genes that influence
metabolism and cell survival. These congenic dissection
studies are important for two reasons. Firstly, the separa
tion between autoantibodies and nephritis can explain the
rare clinical situation of kidney disease in patients with
SLE without appreciable autoantibody titres. Secondly, the
concept of genes that afford protection from target organ
damage changes some of our long-held notions regard
ing the pathogenesis of lupus nephritis, clarifies the large
between-patient variability in disease expression that can
be observed, and has important potential implications for
treatmentrecommendations.146,147

Accessibility of apoptotic debris

The final category of molecules that contribute to the


pathology of SLE comprises genes that could impact
thecirculating or local tissue levels of accessible chro
matin, which is predominantly generated from apop
totic cells and immune complexes. These genes include
DNASE1, TREX1, FcR, ITGAM, C1Q (and related comple
ment components) and ATG5.148150 Although deficien
cies of many of these genes are associated with SLE, the
underlying mechanisms contributing to disease pathology
remain unclear.148 In particular, ATG5 might serve a key
function in podocyte biology, as multiple nephropathies
can be induced when ATG5 gene function is impaired.151
Importantly, podocyte-specific deletion of Atg5 in a
murine model resulted in the accumulation ofubiqui
tinated and oxidized proteins, endoplasmic reticulum
stress, and proteinuria, indicating that autophagy is an
important homeostatic mechanism for maintaining
podocyte function and preventing glomerular injury.151

Exposure of nuclear autoantigens


Previous research has established that nuclear antigens
are targeted in SLE, which is characterized by a strong

8 | ADVANCE ONLINE PUBLICATION

www.nature.com/nrneph
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
serological response to DNA, histones and ribonuclear
proteins.152,153 These nuclear antigens are not usually
exposed to the immune system as they are sequestered
within cellular and nuclear membranes. Consequently,
numerous studies have aimed to uncover the processes
by which nuclear autoantigens become exposed and
drive SLE-associated autoimmune responses.149,150,154
Although several known pathways can lead to cell death
(with more being discovered), apoptosis remains the
dominant mechanism by which cells die. Rapid clearance
of apoptotic cells prevents immunogenicity or the ability
to initiate an inflammatory response. Experimental evi
dence suggests that accumulation of apoptotic debris,
which can occur as a result of failure of this clearance
machinery, is an important contributor to autoimmunity
in SLE.155,156

Clearance of apoptotic cells


Multiple ligands, receptors, opsonins, and other mol
ecule classes are involved in the clearance of apoptotic
cells and other debris associated with the processes
of cell death. Genetic knockout and pharmacological
studies in mice have demonstrated how genes affecting
autoantigen clearance could promote the production of
anti-nuclear autoantibodies and trigger other features
of SLE.149 Nevertheless, few of these genes have thus far
been implicated in SLE among human patients. Some
illustrative examples in murine models include defi
ciency of Tyro3, Axl and Mertk (also known as Tam)
receptors that are important for binding to apoptotic
cells and their subsequent engulfmentthat results
in the development of autoantibodies, arthritis, skin
rashes and glomerular immune complex deposition.157
Mice deficient in T-cell IgG4 (TIM-4), which binds the
phosphatidyl serine residues exposed on the surface
of apoptotic cells, exhibit anti-dsDNA antibodies and
elevated Bcell and Tcell activation.158 Other proteins
that regulate apoptotic cell clearance include MFGE8,
MBL2, APCS, and CRP.149,150
NETosis
NETosis is a specialized form of cell death that occurs
primarily in neutrophils, during which neutrophil
extracellular traps (NETs) are released. NETs comprise
a mesh of DNA and histones, as well as the content of
cytoplasmic granules and other key mediators, such as
HMGB1.159 NETs are becoming increasingly recognized
as a major source of nuclear antigens and other inflam
matory stimuli that can help drive autoimmunity in
SLE.159,160 NETs form more easily in patients with SLE
as compared to healthy controls, and NET clearance is
impaired in patients with SLE,159 perhaps owing to the
presence of anti-DNASE1 or anti-NET antibodies.154
NETs can stimulate dendritic cells to secrete IFNI, and
might also cause direct damage to tissues.159 DNASE1
is essential for the degradation of NETs and hence is
pivotal to their clearance. Mice deficient in Dnase1
develop an SLE-like syndrome, and many patients with
SLE exhibit decreased DNASE1 activity. Furthermore,
genetic mutations in DNASE1 have been linked to SLE

in patients.149,150 Whether any of the genes associated


with susceptibility to SLE or lupus nephritis have an
impact on NETosis remains to be established.
Opsonization
Opsonization of apoptotic cells by serum proteins has a
pivotal role in cell clearance. The opsonin C1Q is addi
tionally involved in the clearance of NETs and has been
closely linked with SLE.161,162 C1q-deficient mice develop
antinuclear autoimmunity, and almost all patients with
C1Q deficiency develop SLE. Moreover, anti-C1Q anti
bodies are frequently found in patients with SLE, which
can induce a functional state of C1Q deficiency and
hinder the clearance of apoptotic cells.163 Other comple
ment proteins, such as C2, C3, and C4, might be neces
sary for phagocytosis; indeed, genetic associations with
SLE have been described not only for C1Q, but also
withmutations in C2, C4, CR3 and complement inhibi
tors.163 Finally, although the association between C1Q
and SLE has been attributed to the clearance of dying
cells, findings from some studies have suggested a role
of complement proteins in several other areas perti
nent to the pathogenesis of SLE, including regulation of
lymphocytes and expression of IFN-I.163

Conclusions

Genetic studies of SLE and lupus nephritis have already


highlighted more than 50 potential candidate genes that
contribute to disease pathology, many of which can be
subdivided into one of four key molecular pathways.
Although many more genes are likely to be identified in
the coming years, the molecular pathways implicated by
these disease genes yield insights into some of the key
steps that might be important for the pathogenesis of
lupus nephritis. Genes that breach immune tolerance
and promote autoantibody production, notably antidsDNA, might act in concert with genes that augment
innate immune signalling and IFN-I production to gen
erate waves of effector leucocyte secretion and release
of inflammatory mediators and autoantibodies that
together initiate renal assault.164166 The presence of
cognate antigens in the glomerular matrix, together with
intrinsic molecular abnormalities in resident renal cells,
might further accentuate the progression of the disease
among patients with lupus nephritis.164,167 Differential
activation of the various molecular pathways discussed
in this Review could conceivably lead to the diverse clini
cal presentations of SLE and lupus nephritis. Deciphering
the molecular basis of disease in each patient would
help physicians to tailor therapy accordingly. Patients
with genetic polymorphisms that lead to activation of
Bcells or Tcells might, for example, benefit from thera
peutics that target specific signalling molecules or co-
stimulatory pathways encoded by the disease genes.
Patients with SLE risk alleles that promote end-organ
inflammation might benefit from therapeutics that target
the implicated pathways within the kidneys.
The field is yet to arrive at such a level of sophisti
cation, where therapies can potentially be tailored to
the genotype of the patient. Firstly, the spectrum of

NATURE REVIEWS | NEPHROLOGY

ADVANCE ONLINE PUBLICATION | 9


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
genes responsible for SLE is still evolving, and the spe
cific genetic aberrations responsible for disease within
each implicated gene are only starting to be elucidated.
Secondly, better biomarkers are required to rapidly clas
sify patients according to the molecular disease pathways
they might harbour. Thirdly, a better armamentarium of
therapeutics is warranted, targeting the respective disease
pathways. Finally, clinical trials need to be conducted to
evaluate if pathway-tailored therapy does indeed reduce
long-term morbidity and mortality in patients with SLE.
Clearly, the next decade will witness intense activity in
all of these quarters.
1.

2.

3.

4.
5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

Feldman, C. H. et al. Epidemiology and


sociodemographics of systemic lupus
erythematosus and lupus nephritis among US
adults with Medicaid coverage, 20002004.
Arthritis Rheum. 65, 753763 (2013).
Rahman, A. & Isenberg, D.A. Systemic lupus
erythematosus. N. Engl. J. Med. 358, 929939
(2008).
Schwartz, N., Goilav, B. & Putterman, C. The
pathogenesis, diagnosis and treatment of lupus
nephritis. Curr. Opin. Rheumatol. 26, 502509
(2014).
Tsokos, G.C. Systemic lupus erythematosus.
N.Engl. J. Med. 365, 21102121 (2011).
Sang, A., Zheng, Y.Y. & Morel, L. Contributions of
Bcells to lupus pathogenesis. Mol. Immunol. 62,
329338 (2014).
Davidson, A. & Aranow, C. Lupus nephritis:
lessons from murine models. Nat. Rev.
Rheumatol. 6, 1320 (2010).
Reddy, V., Jayne, D., Close, D. & Isenberg, D.
Bcell depletion in SLE: clinical and trial
experience with rituximab and ocrelizumab and
implications for study design. Arthritis Res. Ther.
15 (Suppl. 1), S2 (2013).
Liu, Y. & Anders, H.J. Lupus nephritis: from
pathogenesis to targets for biologic treatment.
Nephron Clin. Pract. (2014).
Okamoto, A. et al. Kidney-infiltrating CD4+ Tcell
clones promote nephritis in lupus-prone mice.
Kidney Int. 82, 969979 (2012).
Zhang, Z., Kyttaris, V.C. & Tsokos, G.C. The role
of IL23/IL17 axis in lupus nephritis. J. Immunol.
183, 31603169 (2009).
Miyake, K., Akahoshi, M. & Nakashima, H.
Thsubset balance in lupus nephritis. J. Biomed.
Biotechnol. 2011, 980286 (2011).
Aringer, M., Gunther, C. & Lee-Kirsch, M.A.
Innate immune processes in lupus
erythematosus. Clin. Immunol. 147, 216222
(2013).
Lech, M. & Anders, H.J. The pathogenesis
oflupus nephritis. J. Am. Soc. Nephrol. 24,
13571366 (2013).
Kiefer, K., Oropallo, M.A., Cancro, M.P. &
Marshak-Rothstein, A. Role of typeI interferons
in the activation of autoreactive Bcells.
Immunol. Cell Biol. 90, 498504 (2012).
Bagavant, H. & Fu, S.M. Pathogenesis of kidney
disease in systemic lupus erythematosus. Curr.
Opin. Rheumatol. 21, 489494 (2009).
Nowling, T.K. & Gilkeson, G.S. Mechanisms
oftissue injury in lupus nephritis. Arthritis Res.
Ther. 13, 250 (2011).
Guerra, S.G., Vyse, T.J. & Cunninghame
Graham,D.S. The genetics of lupus:
afunctional perspective. Arthritis Res. Ther.
14,211 (2012).
Flesher, D.L., Sun, X., Behrens, T.W.,
Graham,R.R. & Criswell, L.A. Recent advances

19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

Review criteria
The articles for this Review were selected as follows: we
first identified all genes that have been implicated in SLE
and/or lupus nephritis and validated by further analytical
techniques. Publications relating to these genes and
their reported functions were gathered from PubMed
and Google Scholar using the gene name as a search
term. Additional search terms included lupus, SLE,
nephritis, genetics and GWAS. Further articles were
identified from the reference lists of these publications.
The search was confined to articles published in English,
and all years of publication were included.

in the genetics of systemic lupus erythematosus.


Expert Rev. Clin. Immunol. 6, 461479 (2010).
Crispin, J.C., Hedrich, C.M. & Tsokos, G.C.
Gene-function studies in systemic lupus
erythematosus. Nat. Rev. Rheumatol. 9,
476484 (2013).
Deng, Y. & Tsao, B.P. Genetic susceptibility
tosystemic lupus erythematosus in the
genomicera. Nat. Rev. Rheumatol. 6, 683692
(2010).
Graham, R.R. etal. Genetic variants near
TNFAIP3 on 6q23 are associated with
systemiclupus erythematosus. Nat. Genet. 40,
10591061 (2008).
Guo, Y., Orme, J. & Mohan, C. A genopedia of
lupus genes - lessons from gene knockouts.
Curr. Rheumatol. Rev. 9, 9099 (2013).
Niewold, T.B. etal. IRF5 haplotypes demonstrate
diverse serological associations which predict
serum interferon alpha activity and explain the
majority of the genetic association with systemic
lupus erythematosus. Ann. Rheum. Dis. 71,
463468 (2012).
Jensen, M.A. etal. Functional genetic
polymorphisms in ILT3 are associated with
decreased surface expression on dendritic
cellsand increased serum cytokines in lupus
patients. Ann. Rheum. Dis. 72, 596601 (2013).
Richez, C. etal. IFN regulatory factor 5 is
required for disease development in the
FcgammaRIIB/Yaa and FcgammaRIIB/
mouse models of systemic lupus
erythematosus. J. Immunol. 184, 796806
(2010).
Qin, L. etal. Association of IRF5 gene
polymorphisms and lupus nephritis in a Chinese
population. Nephrology (Carlton) 15, 710713
(2010).
Morel, L. etal. Genetic reconstitution of systemic
lupus erythematosus immunopathology with
polycongenic murine strains. Proc. Natl Acad. Sci.
USA 97, 66706675 (2000).
Mohan, C. et al. Genetic dissection of Sle
pathogenesis: Sle3 on murine chromosome 7
impacts Tcell activation, differentiation, and cell
death. J. Immunol. 162, 64926502 (1999).
Xie, S. & Mohan, C. Divide and conquerthe
power of congenic strains. Clin. Immunol. 110,
109111 (2004).
Henry, T. & Mohan, C. Systemic lupus
erythematosusrecent clues from congenic
strains. Arch. Immunol. Ther. Exp. (Warsz.) 53,
207212 (2005).
Mohan, C. etal. Genetic dissection of lupus
pathogenesis: a recipe for nephrophilic
autoantibodies. J. Clin. Invest. 103, 16851695
(1999).
Morel, L. etal. Functional dissection of systemic
lupus erythematosus using congenic mouse
strains. J. Immunol. 158, 60196028 (1997).

10 | ADVANCE ONLINE PUBLICATION

33. Vaughn, S.E. et al. Genetic susceptibility to


lupus: the biological basis of genetic risk found
in Bcell signaling pathways. J. Leukoc. Biol. 92,
577591 (2012).
34. Avalos, A.M., Meyer-Wentrup, F. & Ploegh, H.L.
Bcell receptor signaling in lymphoid
malignancies and autoimmunity. Adv. Immunol.
123, 149 (2014).
35. Shao, W.H. & Cohen, P.L. The role of tyrosine
kinases in systemic lupus erythematosus and
their potential as therapeutic targets. Expert
Rev. Clin. Immunol. 10, 573582 (2014).
36. Castillejo-Lopez, C. etal. Genetic and physical
interaction of the Bcell systemic lupus
erythematosus-associated genes BANK1 and
BLK. Ann. Rheum. Dis. 71, 136142 (2012).
37. Coughlin, J. J. et al. RasGRP1 and RasGRP3
regulate Bcell proliferation by facilitating Bcell
receptor-Ras signaling. J. Immunol. 175,
71797184 (2005).
38. Shao, B. etal. Hyperglycaemia promotes human
brain microvascular endothelial cell apoptosis
via induction of protein kinase CI and
prooxidant enzyme NADPH oxidase. Redox Biol.
2, 694-701 (2014).
39. Liu, L. etal. PKCII acts downstream of
chemoattractant receptors and mTORC2 to
regulate cAMP production and myosin II activity
in neutrophils. Mol. Biol. Cell 25, 1446145
(2014).
40. Zheng, Y. etal. Phosphorylation of RasGRP3
onthreonine 133 provides a mechanistic link
between PKC and Ras signaling systems in
Bcells. Blood 105, 36483654. (2005).
41. Roberts, D. M. et al. A vascular gene trap screen
defines RasGRP3 as an angiogenesis-regulated
gene required for the endothelial response to
phorbol esters. Mol. Cell Biol. 24, 1051510528
(2004).
42. Oleksyn, D. etal. Protein kinase C is required
for lupus development in Sle mice. Arthritis
Rheum. 65, 10221031 (2013).
43. Yu, C.C., Mamchak, A.A. & DeFranco, A.L.
Signaling mutations and autoimmunity. Curr. Dir.
Autoimmun. 6, 6188 (2003).
44. Lamagna, C. et al. Bcell-specific loss of Lyn
kinase leads to autoimmunity. J. Immunol. 192,
919928 (2014).
45. Hua, Z. etal. Requirement for MyD88 signaling
in Bcells and dendritic cells for germinal center
anti-nuclear antibody production in Lyn-deficient
mice. J. Immunol. 192, 875885 (2014).
46. Yu, C.C., Yen, T.S., Lowell, C.A. & DeFranco,A.L.
Lupus-like kidney disease in mice deficient in the
Src family tyrosine kinases Lyn and Fyn. Curr. Biol.
11, 3438 (2001).
47. Samuelson, E.M. et al. Blk haploinsufficiency
impairs the development, but enhances the
functional responses, of MZ Bcells. Immunol.
Cell Biol. 90, 620629 (2012).

www.nature.com/nrneph
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
48. Samuelson, E.M. etal. Reduced B lymphoid
kinase (Blk) expression enhances
proinflammatory cytokine production and
induces nephrosis in C57BL/6lpr/lpr mice.
PLoS ONE 9, e92054 (2014).
49. Hata, A. et al. Functional analysis of Csk in signal
transduction through the Bcell antigen receptor.
Mol. Cell Biol. 14, 73067313 (1994).
50. Manjarrez-Orduno, N. etal. CSK regulatory
polymorphism is associated with systemic lupus
erythematosus and influences Bcell signaling
and activation. Nat. Genet. 44, 12271230
(2012).
51. Dai, X. etal. A disease-associated PTPN22
variant promotes systemic autoimmunity in
murine models. J. Clin. Invest. 123, 20242036
(2013).
52. Bayley, R. etal. The autoimmune-associated
genetic variant PTPN22 R620W enhances
neutrophil activation and function in patients
with rheumatoid arthritis and healthy individuals.
Ann. Rheum. Dis. http://dx.doi.org/10.1136/
annrheumdis-2013-204796.
53. Zhang, J. etal. The autoimmune diseaseassociated PTPN22 variant promotes calpainmediated Lyp/Pep degradation associated
withlymphocyte and dendritic cell
hyperresponsiveness. Nat. Genet. 43, 902907
(2011).
54. Brownlie, R. J. et al. Lack of the phosphatase
PTPN22 increases adhesion of murine
regulatory T cells to improve their
immunosuppressive function. Sci. Signal. 5, ra87
(2012).
55. Ivashkiv, L. B. PTPN22 in autoimmunity: different
cell and different way. Immunity 25, 9193
(2013).
56. Leadbetter, E.A. etal. Chromatin-IgG complexes
activate Bcells by dual engagement of IgM and
Toll-like receptors. Nature 416, 603607 (2002).
57. Mohan, C., Adams, S., Stanik, V. & Datta, S.K.
Nucleosome: a major immunogen for pathogenic
autoantibody-inducing Tcells of lupus. J. Exp.
Med. 177, 13671381 (1993).
58. Barcellos, L.F. etal. High-density SNP screening
of the major histocompatibility complex in
systemic lupus erythematosus demonstrates
strong evidence for independent susceptibility
regions. PLoS Genet. 5, e1000696 (2009).
59. Graham, R.R. etal. Specific combinations of
HLA-DR2 and DR3 class II haplotypes contribute
graded risk for disease susceptibility and
autoantibodies in human SLE. Eur. J. Hum.
Genet. 15, 823830 (2007).
60. Fernando, M.M. etal. Defining the role of the
MHC in autoimmunity: a review and pooled
analysis. PLoS Genet. 4, e1000024 (2008).
61. Sharpe, A.H. Mechanisms of costimulation.
Immunol. Rev. 229, 511 (2009).
62. Farres, M.N., Al-Zifzaf, D.S., Aly, A.A. &
AbdRaboh, N.M. OX40/OX40L in systemic
lupus erythematosus: association with disease
activity and lupus nephritis. Ann. Saudi Med. 31,
2934 (2011).
64. Shah, K. etal. Dysregulated balance of Th17
andTh1 cells in systemic lupus erythematosus.
Arthritis Res. Ther. 12, R53 (2010).
65. Chang, A. etal. Insitu Bcell-mediated immune
responses and tubulointerstitial inflammation
inhuman lupus nephritis. J. Immunol. 186,
18491860 (2011).
66. Neubert, K. etal. The proteasome inhibitor
bortezomib depletes plasma cells and protects
mice with lupus-like disease from nephritis.
Nat.Med. 14, 748755 (2008).
67. Jacobi, A.M. etal. Correlation between
circulating CD27high plasma cells and disease
activity in patients with systemic lupus

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

85.

erythematosus. Arthritis Rheum. 48, 13321342


(2003).
Yang, W. etal. Genome-wide association study
inAsian populations identifies variants in ETS1
and WDFY4 associated with systemic lupus
erythematosus. PLoS Genet. 6, e1000841
(2010).
Wang, D. etal. Ets1 deficiency leads to altered
Bcell differentiation, hyperresponsiveness to
TLR9 and autoimmune disease. Int. Immunol.
17, 11791191 (2005).
Kim, S.J. et al. Tolerogenic function of Blimp1
indendritic cells. J. Exp. Med. 208, 21932199
(2011).
Zhou, Z. etal. Blimp1 siRNA inhibits Bcell
differentiation and prevents the development
oflupus in mice. Hum. Immunol. 74, 297301
(2013).
Amarilyo, G., Lourenco, E.V., Shi, F.D. &
La Cava, A. IL17 promotes murine lupus.
J.Immunol. 193, 540543 (2014).
Schmidt, T. et al. Function of the Th17/
interleukin17A immune response in murine
lupus nephritis. Arthritis Rheumatol. 67,
475487 (2015).
Tan, W. etal. Association of PPP2CA
polymorphisms with systemic lupus
erythematosus susceptibility in multiple ethnic
groups. Arthritis Rheum. 63, 27552763
(2011).
Lashine, Y.A., Salah, S., Aboelenein, H.R. &
Abdelaziz, A.I. Correcting the expression of
miRNA155 represses PP2Ac and enhances
therelease of IL2 in PBMCs of juvenile SLE
patients. Lupus 24, 240247 (2014).
Crispin, J.C. etal. Cutting edge: protein
phosphatase 2A confers susceptibility
toautoimmune disease through an
IL17dependent mechanism. J. Immunol. 188,
35673571 (2012).
Apostolidis, S.A. et al. Protein phosphatase 2A
enables expression of interleukin 17 (IL17)
through chromatin remodeling. J. Biol. Chem.
288, 2677526784 (2013).
Richman, I.B. etal. European genetic ancestry
is associated with a decreased risk of lupus
nephritis. Arthritis Rheum. 64, 33743382
(2012).
Taylor, K.E. etal. Risk alleles for systemic lupus
erythematosus in a large case-control collection
and associations with clinical subphenotypes.
PLoS Genet. 7, e1001311 (2011).
Taylor, K.E. etal. Specificity of the STAT4 genetic
association for severe disease manifestations
of systemic lupus erythematosus. PLoS Genet.
4, e1000084 (2008).
Jacob, C.O. etal. Pivotal role of Stat4 and Stat6
in the pathogenesis of the lupus-like disease in
the New Zealand mixed 2328 mice. J. Immunol.
171, 15641571 (2003).
Rajabi, P., Alaee, M., Mousavizadeh, K. &
Samadikuchaksaraei, A. Altered expression of
TNFSF4 and TRAF2 mRNAs in peripheral blood
mononuclear cells in patients with systemic
lupus erythematosus: association with
atherosclerotic symptoms and lupus nephritis.
Inflamm. Res. 61, 13471354 (2012).
Zhou, X.J. et al. A replication study from Chinese
supports association between lupus-risk allele
in TNFSF4 and renal disorder. Biomed. Res. Int.
2013, 597921 (2013).
Sanchez, E. etal. Phenotypic associations of
genetic susceptibility loci in systemic lupus
erythematosus. Ann. Rheum. Dis. 70,
17521757 (2011).
Obermoser, G. & Pascual, V. The interferon-alpha
signature of systemic lupus erythematosus.
Lupus 19, 10121019 (2010).

NATURE REVIEWS | NEPHROLOGY

86. Fairhurst, A.M. etal. TypeI interferons


producedby resident renal cells may promote
end-organ disease in autoantibody-mediated
glomerulonephritis. J. Immunol. 183,
68316838 (2009).
87. Triantafyllopoulou, A. etal. Proliferative lesions
and metalloproteinase activity in murine lupus
nephritis mediated by typeI interferons and
macrophages. Proc. Natl Acad. Sci. USA 107,
30123017 (2010).
88. He, C.F. etal. TNIP1, SLC15A4, ETS1, RasGRP3
and IKZF1 are associated with clinical features
of systemic lupus erythematosus in a Chinese
Han population. Lupus 19, 11811186 (2010).
89. Frucht, D. M. etal. STAT4 is expressed in
activated peripheral blood monocytes, dendritic
cells, and macrophages at sites of Th1-mediated
inflammation. J. Immunol. 164, 46594664
(2000).
90. Kaplan, M. H. STAT4: a critical regulator
ofinflammation invivo. Immunol. Res. 31,
231242 (2005).
91. Iwakura, Y. & Ishigame, H. The IL23/IL17 axis
in inflammation. J. Clin. Invest. 116, 12181222
(2006).
92. Ivashkiv, L. B. & Donlin, L. T. Regulation of typeI
interferon responses. Nat. Rev. Immunol. 14,
3649 (2014).
93. Elkon, K. B. & Stone, V. V. TypeI interferon and
systemic lupus erythematosus. J. Interferon
Cytokine Res. 31, 803812 (2011).
94. Dang, J. etal. Gene-gene interactions of IRF5,
STAT4, IKZF1 and ETS1 in systemic lupus
erythematosus. Tissue Antigens 83, 401408
(2014).
95. Karassa, F.B. et al. The Fc gamma RIIIAF158
allele is a risk factor for the development of
lupus nephritis: a meta-analysis. Kidney Int. 63,
14751482 (2003).
96. Brown, E.E., Edberg, J.C. & Kimberly, R.P.
Fcreceptor genes and the systemic lupus
erythematosus diathesis. Autoimmunity 40,
567581 (2007).
97. Floto, R.A. etal. Loss of function of a lupusassociated FcgammaRIIb polymorphism
throughexclusion from lipid rafts. Nat. Med. 11,
10561058 (2005).
98. Breunis, W.B. etal. Copy number variation
attheFCGR locus includes FCGR3A,
FCGRXXXXX2C and FCGR3B but not FCGR2A
andFCGR2B. Hum. Mutat. 30, E640E650
(2009).
99. Fanciulli, M. etal. FCGR3B copy number variation
is associated with susceptibility to systemic, but
not organ-specific, autoimmunity. Nat. Genet. 39,
721723 (2007).
100. Morris, D.L. etal. Evidence for both copy
number and allelic (NA1/NA2) risk at the
FCGR3B locus in systemic lupus erythematosus.
Eur. J. Hum. Genet. 18, 10271031 (2010).
101. Clynes, R., Dumitru, C. & Ravetch, J.V.
Uncoupling of immune complex formation
andkidney damage in autoimmune
glomerulonephritis. Science 279, 10521054
(1998).
102. Bergtold, A. et al. FcR-bearing myeloid cells are
responsible for triggering murine lupus nephritis.
J. Immunol. 177, 72877295 (2006).
103. Snchez-Mejorada, G. & Rosales, C. Signal
transduction by immunoglobulin Fc receptors.
J.Leukoc. Biol. 63, 521533 (1998).
104. Celhar, T., Magalhes, R. & Fairhurst, A. M. TLR7
and TLR9 in SLE: when sensing self goes wrong.
Immunol. Res. 53, 5877 (2012).
105. Shrivastav, M. & Niewold, T. B. Nucleic acid
sensors and typeI interferon production in
systemic lupus erythematosus. Front. Immunol.
4, 319 (2013).

ADVANCE ONLINE PUBLICATION | 11


2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
106. Zhou, X.J. etal. Association of TLR9 gene
polymorphisms with lupus nephritis in a
ChineseHan population. Clin. Exp. Rheumatol.
28, 397400 (2010).
107. Jacob, C.O. etal. Identification of IRAK1 as a
risk gene with critical role in the pathogenesis of
systemic lupus erythematosus. Proc. Natl Acad.
Sci. USA 106, 62566261 (2009).
108. Barrat, F.J. et al. Treatment of lupus-prone mice
with a dual inhibitor of TLR7 and TLR9 leads to
reduction of autoantibody production and
amelioration of disease symptoms. Eur. J.
Immunol. 37, 35823586 (2007).
109. Christensen, S.R. etal. Toll-like receptor 7 and
TLR9 dictate autoantibody specificity and have
opposing inflammatory and regulatory roles in a
murine model of lupus. Immunity 25, 417428
(2006).
110. Verstrepen, L. et al. ABINs: A20 binding
inhibitors of NFkappa B and apoptosis
signaling. Biochem. Pharmacol. 78, 105114
(2009).
111. Lewis, M. J. et al. BE2L3 polymorphism amplifies
NFB activation and promotes plasma cell
development, linking linear ubiquitination to
multiple autoimmune diseases. Am. J. Hum.
Genet. 96, 221234 (2015).
112. Zuo, X. B. et al. Variants in TNFSF4, TNFAIP3,
TNIP1, BLK, SLC15A4 and UBE2L3 interact to
confer risk of systemic lupus erythematosus
inChinese population. Rheumatol. Int. 34,
459464 (2014).
113. Hovelmeyer, N. etal. A20 deficiency in Bcells
enhances Bcell proliferation and results in the
development of autoantibodies. Eur. J. Immunol.
41, 595601 (2011).
114. Caster, D.J. etal. ABIN1 dysfunction as a
genetic basis for lupus nephritis. J. Am. Soc.
Nephrol. 24, 17431754 (2013).
115. Bates, J.S. etal. Meta-analysis and imputation
identifies a 109kb risk haplotype spanning
TNFAIP3 associated with lupus nephritis and
hematologic manifestations. Genes Immun. 10,
470477 (2009).
116. Zhou, Y. et al. Multiple lupus-associated ITGAM
variants alter Mac1 functions on neutrophils.
Arthritis Rheum. 65, 29072916 (2013).
117. Fagerholm, S. C. et al. The CD11b-integrin
(ITGAM) and systemic lupus erythematosus.
Lupus 22, 657663 (2013).
118. Ross, G. D. & Ve
tvicka, V. CR3 (CD11b, CD18):
aphagocyte and NK cell membrane receptor
with multiple ligand specificities and functions.
Clin. Exp. Immunol. 92, 181184 (1993).
119. Kim-Howard, X. etal. ITGAM coding variant
(rs1143679) influences the risk of renal
disease, discoid rash and immunological
manifestations in patients with systemic lupus
erythematosus with European ancestry. Ann.
Rheum. Dis. 69, 13291332 (2010).
120. Yang, W. etal. ITGAM is associated with disease
susceptibility and renal nephritis of systemic
lupus erythematosus in Hong Kong Chinese and
Thai. Hum. Mol. Genet. 18, 20632070 (2009).
121. Orme, J. & Mohan, C. Macrophages and
neutrophils in SLE-An online molecular catalog.
Autoimmun. Rev. 11, 365372 (2012).
122. Crispn, J. C., Hedrich, C. M. & Tsokos, G. C.
Gene-function studies in systemic lupus
erythematosus. Nat. Rev. Rheumatol. 9,
476484 (2013).
123. Seredkina, N. et al. Lupus nephritis: enigmas,
conflicting models and an emerging concept.
Mol. Med. 19, 161169 (2013).
124. Rekvig, O. P. & Van der Vlag, J. The pathogenesis
and diagnosis of systemic lupus erythematosus:
still not resolved. Semin. Immunopathol. 36,
301311 (2014).

125. Zhou, T.B. etal. Relationship between


angiotensin-converting enzyme insertion/
deletion gene polymorphism and systemic lupus
erythematosus/lupus nephritis: a systematic
review and metaanalysis. J. Rheumatol. 39,
686693 (2012).
126. Chung, S.A. etal. Differential genetic
associations for systemic lupus erythematosus
based on anti-dsDNA autoantibody production.
PLoS Genet. 7, e1001323 (2011).
127. Liu, K. etal. Kallikrein genes are associated
withlupus and glomerular basement membranespecific antibody-induced nephritis in mice and
humans. J. Clin. Invest. 119, 911923 (2009).
128. Ciferska, H. etal. Expression of nucleic acid
binding Toll-like receptors in control, lupus and
transplanted kidneys-a preliminary pilot study.
Lupus 17, 580585 (2008).
129. Papadimitraki, E.D. et al. Glomerular expression
of toll-like receptor9 in lupus nephritis but not in
normal kidneys: implications for the amplification
of the inflammatory response. Lupus 18,
831835 (2009).
130. Patole, P.S. etal. Expression and regulation of
Toll-like receptors in lupus-like immune complex
glomerulonephritis of MRL-Fas(lpr) mice. Nephrol.
Dial. Transplant. 21, 30623073 (2006).
131. Ka, S.M. etal. Mesangial cells of lupus-prone
mice are sensitive to chemokine production.
Arthritis Res. Ther. 9, R67 (2007).
132. Liu, S. etal. TRAF6 knockdown promotes
survival and inhibits inflammatory response to
lipopolysaccharides in rat primary renal proximal
tubule cells. Acta Physiol. (Oxf.) 199, 339346
(2010).
133. Benigni, A. etal. Involvement of renal tubular
Toll-like receptor 9 in the development of
tubulointerstitial injury in systemic lupus.
Arthritis Rheum. 56, 15691578 (2007).
134. Machida, H. etal. Expression of Toll-like
receptor9 in renal podocytes in childhood-onset
active and inactive lupus nephritis. Nephrol. Dial.
Transplant. 25, 25302537 (2010).
135. Frieri, M. etal. Toll-like receptor 9 and vascular
endothelial growth factor levels in human
kidneys from lupus nephritis patients. J. Nephrol.
25, 10411046 (2012).
136. Lichtnekert, J. etal. Trif is not required for
immune complex glomerulonephritis: dying cells
activate mesangial cells via Tlr2/Myd88 rather
than Tlr3/Trif. Am. J. Physiol. Renal Physiol. 296,
F867F874 (2009).
137. Kunter, U. etal. Combined expression of A1
andA20 achieves optimal protection of renal
proximal tubular epithelial cells. Kidney Int. 68,
15201532 (2005).
138. Lutz, J. etal. The A20 gene protects kidneys
from ischaemia/reperfusion injury by
suppressing pro-inflammatory activation.
J. Mol. Med. (Berl.) 86, 13291339 (2008).
139. da Silva, C.G. etal. Hepatocyte growth factor
preferentially activates the anti-inflammatory
arm of NFB signaling to induce A20 and protect
renal proximal tubular epithelial cells from
inflammation. J. Cell Physiol. 227, 13821390
(2012).
140. Reiser, J. etal. Induction of B71 in podocytes
isassociated with nephrotic syndrome. J. Clin.
Invest. 113, 13901397 (2004).
141. Shimada, M. etal. Toll-like receptor 3 ligands
induce CD80 expression in human podocytes
viaan NFBdependent pathway. Nephrol. Dial.
Transplant. 27, 8189 (2012).
142. Ishimoto, T. etal. Toll-like receptor 3 ligand,
polyIC, induces proteinuria and glomerular
CD80, and increases urinary CD80 in mice.
Nephrol. Dial. Transplant. 28, 14391446
(2013).

12 | ADVANCE ONLINE PUBLICATION

143. Yu, C.C. etal. Abatacept in B71positive


proteinuric kidney disease. N. Engl. J. Med. 369,
24162423 (2013).
144. Ichinose, K. etal. Cutting edge: calcium/
calmodulin-dependent protein kinase typeIV is
essential for mesangial cell proliferation and
lupus nephritis. J. Immunol. 187, 55005504
(2011).
145. Fu, S.M., Deshmukh, U.S. & Gaskin, F.
Pathogenesis of systemic lupus erythematosus
revisited 2011: end organ resistance to damage,
autoantibody initiation and diversification, and
HLA-DR. J. Autoimmun. 37, 104112 (2011).
146. Ge, Y. etal. Cgnz1 allele confers kidney
resistance to damage preventing progression
ofimmune complex-mediated acute lupus
glomerulonephritis. J. Exp. Med. 210,
23872401 (2013).
147. Dai, C. etal. Genetics of systemic lupus
erythematosus: immune responses and
endorgan resistance to damage. Curr. Opin.
Immunol. 31, 8796 (2014).
148. Guo, Y., Orme, J. & Mohan, C. A genopedia of
lupus genes - lessons from gene knockouts.
Curr. Rheumatol. Rev. 9, 9099 (2013).
149. Shao, W.H. & Cohen, P.L. Disturbances of
apoptotic cell clearance in systemic lupus
erythematosus. Arthritis Res. Ther. 13, 202
(2011).
150. Pieterse, E. & Van der Vlag, J. Breaking
immunological tolerance in systemic lupus
erythematosus. Front. Immunol. 5, 164 (2014).
151. Hartleben, B. etal. Autophagy influences
glomerular disease susceptibility and maintains
podocyte homeostasis in aging mice. J. Clin.
Invest. 120, 10841096 (2010).
152. Yaniv, G. etal. A volcanic explosion of
autoantibodies in systemic lupus erythematosus:
a diversity of 180 different antibodies found in
SLE patients. Autoimmun. Rev. 14, 7579 (2015).
153. Mehra, S. & Fritzler, M.J. The spectrum of antichromatin/nucleosome autoantibodies:
independent and interdependent biomarkers of
disease. J. Immunol. Res. 2014, 368274 (2014).
154. Gatto, M. etal. Emerging and critical issues in
the pathogenesis of lupus. Autoimmun. Rev. 12,
523536 (2013).
155. Rekvig, O.P. & Van der Vlag, J. The pathogenesis
and diagnosis of systemic lupus erythematosus:
still not resolved. Semin. Immunopathol. 36,
301311 (2014).
156. Munoz, L.E. et al. The role of defective clearance
of apoptotic cells in systemic autoimmunity. Nat.
Rev. Rheumatol. 6, 280289 (2010).
157. Cohen, P.L. etal. Delayed apoptotic cell
clearance and lupus-like autoimmunity in mice
lacking the cmer membrane tyrosine kinase.
J.Exp. Med. 196, 135140 (2002).
158. Rodriguez-Manzanet, R. et al. T and Bcell
hyperactivity and autoimmunity associated with
niche-specific defects in apoptotic body
clearance in TIM4deficient mice. Proc. Natl
Acad. Sci. USA 107, 87068711 (2010).
159. Knight, J.S. & Kaplan, M.J. Lupus neutrophils:
NET gain in understanding lupus pathogenesis.
Curr. Opin. Rheumatol. 24, 441450 (2012).
160. Magna, M. & Pisetsky, D.S. The role of HMGB1
in the pathogenesis of inflammatory and
autoimmune diseases. Mol. Med. 20, 138146
(2014).
161. Cunninghame Graham, D.S., Akil, M. & Vyse,T.J.
Association of polymorphisms across the
tyrosine kinase gene, TYK2 in UK SLE families.
Rheumatology (Oxford) 46, 927930 (2007).
162. Mevorach, D. Clearance of dying cells and
systemic lupus erythematosus: the role of C1q
and the complement system. Apoptosis 15,
11141123 (2010).

www.nature.com/nrneph
2015 Macmillan Publishers Limited. All rights reserved

REVIEWS
163. Leffler, J., Bengtsson, A.A. & Blom, A.M.
Thecomplement system in systemic lupus
erythematosus: an update. Ann. Rheum. Dis. 73,
16011606 (2014).
164. Saxena, R., Mahajan, T. & Mohan, C. Lupus
nephritis: current update. Arthritis Res. Ther. 13,
240 (2011).
165. Liu, Y. & Anders, H. J. Lupus nephritis: from
pathogenesis to targets for biologic treatment.
Nephron Clin. Pract. 128, 224231 (2014).
166. Liu, Z. & Davidson, A. Taming lupusa new
understanding of pathogenesis is leading to
clinical advances. Nat. Med. 18, 871882
(2012).
167. Putterman, C. New approaches to the renal
pathogenicity of anti-DNA antibodies in systemic
lupus erythematosus. Autoimmun. Rev. 2, 711
(2004).
168. Kozyrev, S.V. etal. Functional variants in the
Bcell gene BANK1 are associated with systemic
lupus erythematosus. Nat. Genet. 40, 211216
(2008).
169. Yang, W. etal. Genome-wide association study
inAsian populations identifies variants in ETS1
and WDFY4 associated with systemic lupus
erythematosus. PLoS Genet. 6, e1000841
(2010).
170. Sanchez, E. etal. Identification of novel genetic
susceptibility loci in African American lupus
patients in a candidate gene association
study.Arthritis Rheum. 63, 34933501
(2011).
171. Hom, G. etal. Association of systemic lupus
erythematosus with C8orf13-BLK and ITGAMITGAX. N. Engl. J. Med. 358, 900909 (2008).
172. Han, J.W. etal. Genome-wide association
studyin a Chinese Han population identifies
nine new susceptibility loci for systemic lupus
erythematosus. Nat. Genet. 41, 12341237
(2009).
173. Guthridge, J.M. etal. Two functional lupusassociated BLK promoter variants control
celltype and developmentalstagespecific
transcription. Am. J. Hum. Genet. 94, 586598
(2014).
174. Okada, Y. etal. A genome-wide association study
identified AFF1 as a susceptibility locus for
systemic lupus eyrthematosus in Japanese.
PLoS Genet. 8, e1002455 (2012).
175. Lee, H.S. etal. Ethnic specificity of lupusassociated loci identified in a genome-wide
association study in Korean women. Ann.
Rheum. Dis. 73, 12401245 (2014).
63. Yang, W. etal. Meta-analysis followed by
replication identifies loci in or near CDKN1B,
TET3, CD80, DRAM1, and ARID5B as associated
with systemic lupus erythematosus in Asians.
Am. J. Hum. Genet. 92, 4151 (2013).
176. Liu, P. etal. IL10 gene polymorphisms and
susceptibility to systemic lupus erythematosus:
a meta-analysis. PLoS One 8, e69547 (2013).
177. Gateva, V. etal. A large-scale replication study
identifies TNIP1, PRDM1, JAZF1, UHRF1BP1
andIL10 as risk loci for systemic lupus
erythematosus. Nat. Genet. 41, 12281233
(2009).
178. Lu, R. etal. Genetic associations of LYN with
systemic lupus erythematosus. Genes Immun.
10, 397403 (2009).
179. Zhou, X.J. etal. Genetic association of PRDM1ATG5 intergenic region and autophagy with
systemic lupus erythematosus in a Chinese
population. Ann. Rheum. Dis. 70, 13301337
(2011).
180. Sheng, Y.J. etal. Follow-up study identifies two
novel susceptibility loci PRKCB and 8p11.21 for

systemic lupus erythematosus. Rheumatology


(Oxford) 50, 682688 (2011).
181. Vang, T. etal. Autoimmune-associated lymphoid
tyrosine phosphatase is a gainoffunction
variant. Nat. Genet. 37, 13171319 (2005).
182. Arechiga, A.F. etal. Cutting edge: the PTPN22
allelic variant associated with autoimmunity
impairs Bcell signaling. J. Immunol. 182,
33433347 (2009).
183. Menard, L. etal. The PTPN22 allele encoding
anR620W variant interferes with the removal
ofdeveloping autoreactive Bcells in humans.
J.Clin. Invest. 121, 363544 (2011).
184. Wang, C. etal. Genes identified in Asian
SLEGWASs are also associated with SLE in
Caucasian populations. Eur. J. Hum. Genet. 21,
994999 (2013).
185. Namjou, B. etal. High-density genotyping of STAT4
reveals multiple haplotypic associations with
systemic lupus erythematosus in different racial
groups. Arthritis Rheum. 60, 10851095 (2009).
186. Harley, J.B. etal. Genome-wide association scan
in women with systemic lupus erythematosus
identifies susceptibility variants in ITGAM, PXK,
KIAA1542 and other loci. Nat. Genet. 40,
204210 (2008).
187. Manku, H. etal. Trans-ancestral studies fine
mapthe SLE-susceptibility locus TNFSF4.
PLoSGenet. 9, e1003554 (2013).
188. Molineros, J.E. etal. Admixture mapping in
lupus identifies multiple functional variants
within IFIH1 associated with apoptosis,
inflammation, and autoantibody production.
PLoS Genet. 9, e1003222 (2013).
189. Cunninghame Graham, D.S. etal. Association
ofNCF2, IKZF1, IRF8, IFIH1, and TYK2 with
systemic lupus erythematosus. PLoS Genet. 7,
e1002341 (2011).
190. Westra, H.J. etal. Systematic identification
oftrans eQTLs as putative drivers of known
disease associations. Nat. Genet. 45,
12381243 (2013).
191. Kaufman, K.M. etal. Fine mapping of Xq28:
both MECP2 and IRAK1 contribute to risk for
systemic lupus erythematosus in multiple
ancestral groups. Ann. Rheum. Dis. 72,
437444 (2013).
192. Koelsch, K.A. etal. Functional characterization
of the MECP2/IRAK1 lupus risk haplotype in
human Tcells and a human MECP2 transgenic
mouse. J. Autoimmun. 41, 168174 (2013).
193. Graham, R.R. etal. A common haplotype of
interferon regulatory factor 5 (IRF5) regulates
splicing and expression and is associated
withincreased risk of systemic lupus
erythematosus. Nat. Genet. 38, 550555 (2006).
194. Graham, R.R. etal. Three functional variants
ofIFN regulatory factor 5 (IRF5) define risk and
protective haplotypes for human lupus. Proc.
Natl Acad. Sci. USA 104, 67586763 (2007).
195. Sigurdsson, S. etal. Comprehensive evaluation
of the genetic variants of interferon regulatory
factor 5 (IRF5) reveals a novel 5bp length
polymorphism as strong risk factor for systemic
lupus erythematosus. Hum. Mol. Genet. 17,
872881 (2008).
196. Lofgren, S.E. etal. Promoter insertion/deletion
in the IRF5 gene is highly associated with
susceptibility to systemic lupus erythematosus
in distinct populations, but exerts a modest
effect on gene expression in peripheral blood
mononuclear cells. J. Rheumatol. 37, 574578
(2010).
197. Fu, Q. etal. Association of a functional IRF7
variant with systemic lupus erythematosus.
Arthritis Rheum. 63, 749754 (2011).

NATURE REVIEWS | NEPHROLOGY

198. Salloum, R. etal. Genetic variation at the IRF7/


PHRF1 locus is associated with autoantibody
profile and serum interferon-alpha activity in
lupus patients. Arthritis Rheum. 62, 553561
(2010).
199. Baccala, R. etal. Essential requirement for IRF8
and SLC15A4 implicates plasmacytoid dendritic
cells in the pathogenesis of lupus. Proc. Natl
Acad. Sci. USA 110, 29402945 (2013).
200. Lessard, C.J. etal. Identification of IRF8,
TMEM39A, and IKZF3-ZPBP2 as susceptibility
loci for systemic lupus erythematosus in a largescale multiracial replication study. Am. J. Hum.
Genet. 90, 648660 (2012).
201. Shen, N. etal. Sex-specific association of
Xlinked Toll-like receptor 7 (TLR7) with male
systemic lupus erythematosus. Proc. Natl Acad.
Sci. USA 107, 1583815843 (2010).
202. Deng, Y. etal. MicroRNA3148 modulates
allelicexpression of toll-like receptor 7 variant
associated with systemic lupus erythematosus.
PLoS Genet. 9, e1003336 (2013).
203. Zhang, J. et al. Association study of TLR9
polymorphisms and systemic lupus
erythematosus in northern Chinese Han
population. Gene 533, 385388 (2014).
204. Wang, S. et al. An enhancer element harboring
variants associated with systemic lupus
erythematosus engages the TNFAIP3 promoter
to influence A20 expression. PLoS Genet. 9,
e1003750 (2013).
205. Agik, S. etal. The autoimmune disease risk
allele of UBE2L3 in African American patients
with systemic lupus erythematosus: a recessive
effect upon subphenotypes. J. Rheumatol. 39,
738 (2012).
206. Wang, S. etal. A functional haplotype of UBE2L3
confers risk for systemic lupus erythematosus.
Genes Immun. 13, 380387 (2012).
207. Shin, H.D. etal. Common DNase I polymorphism
associated with autoantibody production among
systemic lupus erythematosus patients. Hum.
Mol. Genet. 13, 23432350 (2004).
208. Han, S. etal. Evaluation of imputation-based
association in and around the integrinalphaM
(ITGAM) gene and replication of robust
association between a non-synonymous
functional variant within ITGAM and systemic
lupus erythematosus (SLE). Hum. Mol. Genet.
18, 11711180 (2009).
209. Rhodes, B. etal. The rs1143679 (R77H) lupus
associated variant of ITGAM (CD11b) impairs
complement receptor 3 mediated functions
inhuman monocytes. Ann. Rheum. Dis. 71,
20282034 (2012).
210. Fossati-Jimack, L. etal. Phagocytosis is the main
CR3-mediated function affected by the lupusassociated variant of CD11b in human myeloid
cells. PLoS One 8, e57082 (2013).
211. Namjou, B. etal. Evaluation of the TREX1 gene
ina large multi-ancestral lupus cohort. Genes
Immun. 12, 270279 (2011).
Acknowledgements
The authors would like to acknowledge the editorial
assistance of Simanta Pathak, University of Houston,
USA. Relevant studies in Dr. Puttermans laboratory
were supported by grants from the NIH, DK090319
and AR048692. Dr. Putterman is currently a Weston
Visiting Professor at the Weizmann Institute.
Author contributions
Both authors researched the data for the article,
provided substantial contributions to discussions of
its content, wrote the article, and undertook review
and/or editing of the manuscript before submission.

ADVANCE ONLINE PUBLICATION | 13


2015 Macmillan Publishers Limited. All rights reserved

You might also like