Cytokines: Names and Numbers You Should Care About: Stephen R. Holdsworth and Poh-Yi Gan
Cytokines: Names and Numbers You Should Care About: Stephen R. Holdsworth and Poh-Yi Gan
Cytokines: Names and Numbers You Should Care About: Stephen R. Holdsworth and Poh-Yi Gan
Abstract
Cytokines play an important role in host defense against microorganisms. They orchestrate innate immunity by
inducing protective local inflammation and systemic acute phase responses. Cytokines are important in initiating,
amplifying, directing, mediating, and regulating adaptive immunity. Unfortunately, they may also direct tissue
damage if excessive responses occur or if they are involved in directing and mediating autoimmunity. Under these *Center for
circumstances, cytokines are potential therapeutic targets. Over the last 20 years, we have seen the successful Inflammatory
development and clinical implementation of biologic strategies that target key cytokines in specific inflammatory Diseases, Department
diseases with efficacy, specificity, and toxicity profiles challenging conventional drug therapies. These therapies of Medicine, Monash
University, Clayton,
are finding new applications and many new agents show promise. Unfortunately, these new cytokine-based Victoria, Australia;
therapies have had little effect on renal disease. This review provides evidence that common renal diseases, and †Department of
including those causing AKI and the autoimmune proliferative and crescentic forms of GN, have cytokine Nephrology, Monash
mediation profiles that suggest they would be susceptible to cytokine-targeting therapeutic strategies. Health, Clayton,
Victoria, Australia
Clin J Am Soc Nephrol 10: 2243–2254, 2015. doi: 10.2215/CJN.07590714
Correspondence:
Prof. Stephen R.
Introduction colony stimulating factors (granulocyte colony–stimulating Holdsworth, Centre
Cytokines amplify and direct the generation of ap- factor [G-CSF] and granulocyte–macrophage colony– for Inflammatory
propriate patterns of immunity to combat particular stimulating factor [GM-CSF]), and some growth factors Diseases, Department
of Medicine, Monash
microbial threats. These same cytokines can cause host (TGF-b and PDGF). University, Monash
tissue injury if the activation/amplification of host Medical Centre, 246
defense is overexuberant, as occurs in some infective Clayton Road,
and sterile forms of inflammation. These pathologic Clayton, VIC 3168,
Cytokines in Host Defense Australia. E-mail:
cytokine-driven outcomes are seen in many types of Innate Immunity Stephen.holdsworth@
AKI caused by physical, drug, chemical, and ischemic Most living organisms rely on innate immunity (in monash.edu
injury. If immune tolerance is lost and host tissue the absence of adaptive immunity) for host defense.
antigens become autoimmune targets, cytokines can Cytokines play critical roles in orchestrating the rapid
direct and mediate inflammatory autoimmune dis- effective response of leukocytes and parenchymal
eases. Important renal examples are the autoimmune cells to the detection of microorganisms or significant
forms of inflammatory/crescentic GN. Cytokines act in noninfective damage to parenchymal cells. These cells
concert to generate inflammation in host defense and are hardwired with receptors that recognize and re-
disease, but some cytokines attenuate inflammation and spond to common pathogen proteins through Toll-like
induce repair. Individual cytokines can be inhibited by receptors (TLRs) and danger-associated molecular pat-
antibodies or competitive receptors or by the therapeu- tern receptors (1). Although leukocytes are the major
tic use of immunomodulatory cytokines. These biologic source of innate cytokine responses, parenchymal cells
agents are now widely used to treat inflammatory are increasingly recognized as also producing innate
diseases. There are a number of common renal diseases inflammatory cytokines and interacting with leuko-
that could potentially be treated by targeting cytokines. cytes to optimize cytokine responses in generating
This review will address these issues. host defense. The major acute innate cytokines, IL-1,
TNF-a, IL-6, IL-12, CXCL8 (formerly IL-18), G-CSF,
Cytokine Nomenclature and GM-CSF, are used locally to activate endothelial
Cytokines are glycoproteins that regulate the functions cells and local tissue leukocytes (mast cells [MCs], den-
of the immune system. Definitions are imprecise because dritic cells [DCs], gd T cells, and neurones), triggering
of redundancy of function and the capacity of tissue cytokine-mediated amplification loops generating che-
parenchymal cells and leukocytes to produce them. mokine release, generating endothelial cell adhesion
Hence the terms lymphokine and monokine have been molecule expression, slowing blood flow, and increas-
dropped. Originally described by their perceived major ing vascular permeability. These changes facilitate the
function, the term IL has been adopted. When an agreed accumulation of humoral defense proteins, comple-
characterization of a cytokine is broadly accepted, a ment, coagulation proteins, acute phase proteins, and
number is attributed (e.g., IL-6). However, the use of Ig. They also recruit and activate a range of leukocytes
descriptive names for some key cytokines persists, in- (including innate lymphocytes, gd T cells, natural killer
cluding IFNs (a, b, and g), TNF (TNF-a and TNF-b), cells, natural killer T cells [NKT] cells, and innate
www.cjasn.org Vol 10 December, 2015 Copyright © 2015 by the American Society of Nephrology 2243
2244 Clinical Journal of the American Society of Nephrology
leukocyte-like cells). The net result of these orchestrated transcription factors STAT6 and GATA3 producing IL-4,
events is inflammation. Concurrently, two further processes IL-5, IL-9, and IL-13 that drives humoral and IgE mediated
are initiated: a cohort of cytokines (IL-1, IL-6, and TNF-a) are immunity. In 2005, a new distinct Th subset was defined on
generated that act systemically to prepare the whole organ- the basis of its predominant production of IL-17, named
ism for microbial defense by initiating the acute response Th17 cells (4). Lineage commitment of Th17 cells requires
syndrome (2) and local foreign material is processed and TGF-b and IL-6 for the expression of the transcription fac-
presented by antigen-presenting cells (APCs) to initiate tors, STAT3 and RORg-t, whereas maintenance and expan-
adaptive immunity. These secondary processes are also crit- sion of Th17 cells relies on IL-23. Activated Th17 cells
ically dependent on cytokine direction and are also respon- produce IL-17A-F, IL-21, IL-23, and IL-22 and activate cells,
sive to inhibitory cytokine modulation (Figure 1). particularly neutrophils, important in host defense against
extracellular pathogens. Antigen presentation by TGF-b
alone activates Foxp3-inducing regulatory T cells (Tregs),
Adaptive Immunity
CD41 T cells play a central role in adaptive immunity producing IL-10, TGF-b, and IL-35. Tregs play an impor-
which is characterized by antigen specificity and memory/ tant role in modulating effector T-cell responses and pre-
recall capacity. T-cell specificity is a consequence of the di- venting autoimmunity.
versity of T cell receptors (TCRs) from which thymic pro- Three further Th subsets have been defined: Th9, Th22,
cessing eliminates potentially autoreactive T cells to form the and T follicular helper. The Th2 subset is reprogrammed to
T-cell repertoire. Antigen recognition (signal 1) is initiated by become Th9 when IL-4 plus TGF-b activates STAT6, IRF4,
TCR binding to antigens ingested, processed, and presented GATA3, and PU.1 to produce IL-9, IL-10, IL-17, IL-21, and
(as peptides) on major histocompatibility complex molecules IL-22, promoting tissue inflammation (5). Th22 is important
by specialized APCs. Activation of T cells requires secondary in skin immunity (protection and regeneration). TGF-a and
signals provided by costimulatory molecules (including IL-6 activates transcription factor, AHR, to direct the differ-
CD40, CD80, and CD86) expressed on APCs to engage entiation of IL-22, producing Th22 cells. T follicular helper
CD154 or CD28 on T cells. Innate cytokines released at cells migrate to follicular B-cell zones via CXCR5 where they
the time of antigen presentation (a third signal) deter- help B cells activating Bcl-6 to produce IL-21 (6) (Table 1).
mine the specific Th subset differentiation pathway the
(now activated) T cells will follow. In 1986, Mossman and Cytokines as Therapeutic Targets
Coffman demonstrated that there were two separate path- Many conventional immunomodulating drugs induce their
ways of Th subset differentiation, Th1 and Th2 (3). Th1 therapeutic effects, at least in part, by attenuating the actions
lineage commitment is directed by IL-12, which induces the of injurious cytokines. These drugs include glucocorticoids.
specific signal transducers and activators of transcription Their targets include transcription factors, nuclear factor-k B,
(STAT) factors, STAT4 and T-bet, resulting in the production and activator protein 1, inducing transcription of inflamma-
of effector cytokines, IFN-g, and TNF-a by the differentiated tory cytokines (7). They also effect post-translational events,
Th1 cells. Th1 cells activate macrophages to mount cell- including intracellular signaling and effector cytokine mRNA
mediated immune responses against intracellular pathogens. stability (8). Downstream effects reduce leukocyte trafficking
Th2 differentiation occurs in the presence of IL-4 to activate (by attenuating TNF-a– and IL-1b–enhanced endothelial cell
Figure 1. | Production of major acute innate cytokines involved in local and systemic responses following leukocyte activation via TLRs or
danger-associated molecular pattern receptors. DC, dendritic cell; GM-CSF, granulocyte-macrophage colony–stimulating factor; MC, mast
cell; Rc, receptor; TLR, Toll-like receptor.
Clin J Am Soc Nephrol 10: 2243–2254, December, 2015 Cytokine: Names and Numbers, Holdsworth and Gan 2245
B cell maturation
circulating T cells and inhibit IL-2 production. Th cell differ-
CXCR4, CXCR5
TFH entiation shows a shift to Th2 with attenuation of monocyte
IL-12 without effecting IL-10 production. This results in the
IL-21, IL-6
reduction of Th1 responses favoring Th17 (9). A number of
other well established anti-inflammatory drugs (pentoxifylline
IL-21
Bcl-6
and thalidomide) also attenuate inflammatory cytokine gene
transcription (10).
Skin homeostasis
and pathology
CCR4, CCR6,
Manipulation
CCR10
production
and mucus
TGF-b, IL-4
GATA3
IL-22, IL-23
CCR4, CCR6
IgE immunity
STAT6, GATA3
Cell mediated
IFN-g, TNF-a
STAT4, T-bet
immunity
Th1
IL-10, TGF-b,
Transcription factors
Effector responses
IL-6
cytokines
the effects of cytokine-induced cell activation and conse- clinical data available do not support this role in human
quent pathologic inflammation (34). Tofacitinib preferen- RA and IBD. However, preliminary data in MS, targeting
tially inhibits JAK-1 and JAK-3. In clinical trials, the degree IL-17, shows evidence of benefit. Although much more data
of benefit in resistant RA was similar in efficacy to are necessary before firm conclusions can be drawn, these
adalimumab, a TNF-a inhibitor. It may lack specificity be- observations on the therapeutic benefit on inhibiting single
cause side effects, including sepsis, disturbed liver func- cytokines suggest that different combinations of cytokine
tion tests, raised creatinine, and neutropenia, were may direct specific patterns of disease (Table 3).
reported during its use (35).
AKI (46), but IL-1b2/2 mice are not (47). The role of IL-6 in the IFN-a signature (57). The levels of serum IFN-a and
AKI is complex. Evidence in ischemic AKI is consistent with expression of IFN-a–regulated genes correlate with disease
an injurious role for an endogenous IL-6 (48). However in an activity, autoantibodies, and complement levels (58). IFN-a
HgCl2 model, it was shown that while IL-6–mediated inflam- signaling pathway polymorphisms have been shown in fam-
matory responses contributed to injury, IL-6 trans-signaling ilies with SLE (59). IFN-a is thought to be a promising ther-
induced protective responses (49). apeutic target for SLE, and several mAb inhibitors are in
Cytokine-based immunomodulation can potentially be clinical trials. Sifalimumab is a humanized anti–IFN-a
used as preventative or therapeutic in AKI. The therapeutic mAb. Its use in SLE was associated with reduction in SLE
potential of administering anti-inflammatory cytokine IL-10 flares and activity (60). In a recent clinical trial, use of
has been demonstrated to be effective in both ischemic and rontalizumab (another anti–IFN-a mAb) reduced the expres-
cisplatin-induced AKI (50). More recently, it has been appre- sion patterns of IFN-a–driven genes, improved disease se-
ciated that Tregs can protect from cisplatin (51) and ischemic verity, and improved flare rates in patients with SLE.
AKI (52). Adoptive transfer of Tregs before cisplatin and However, patients with lupus nephritis were not included
before ischemia protected from the development of AKI. in this trial. AGS-009 is an IgG4 humanized mAb that in-
The number of Tregs required for protection in mice sug- duced significant attenuation of IFN-a signatures after a
gests the procedure is feasible in humans. Additionally, single dose (61). A novel approach has been to vaccinate
transferring Tregs to mice 24 hours after ischemic AKI was patients with SLE with IFN-a–kinoid molecules to induce
beneficial in promoting repair (53). autoantibodies to IFN-a. All immunized patients returned
Cytokines released from kidneys with AKI can have the IFN-a signature to baseline (62).
significant effects on distal organs by circulatory spillover. TNF-a. Mouse models of lupus nephritis have shown
Mortality in intensive care units is predicted by distal both potentially protective and accentuating roles for TNF-a.
organ involvement in AKI. Recent studies suggest systemic New Zealand Black (NZB)/New Zealand White (NZW) and
proinflammatory effects are triggered in three waves by the MRL/lpr mice with decreased synthetic capacity of TNF-a
immune release of host alarm signals (alarmins) from the develop lupus nephritis, but in the kidney, intrarenal ex-
AKI-damaged kidney (54). This begins with a uric acid surge, pression of TNF-a correlates with disease activity and in-
which induces a second wave of endothelial cell Weibel– flammation (63). In NZB/NZW mice with IFN-a–induced
Palade bodies released, which is then followed by a third nephritis, anti–TNF-a antibodies attenuate renal inflam-
wave of high-mobility group box 1 protein release. These mation and injury despite maintained immune complex
events are potent triggers for GM-CSF, IFN-g, CXCL8, deposition (64).
G-CSF, IL-12, TNF-a, and IL-6 (55). Recently, renal DCs The data on the role of TNF-a is conflicting; hence, the
were implicated in inducing cytokine-mediated injury in benefits of targeting TNF-a in human lupus nephritis is un-
ischemic AKI. Renal DCs can powerfully influence AKI by certain. However, there is the general view that there is suf-
enhancing or attenuating injury. After injury, DCs initiate ficient data to advise against TNF-a inhibition. Several studies
innate inflammatory responses presenting glycolipids and show that circulating levels of TNF-a and renal expression is
stimulating NKT cells, recruiting neutrophils and initiating increased (65); however, other studies show TNF-a produc-
the IL-17/IL-23 signaling pathway. DCs produce TNF-a, tion by PBMCs was lower in patients with lupus nephritis
IL-6, IL-12, IL-23, IL-17, and IFN-g to amplify injury and than controls (66). Additionally, TNF-a production was as-
inflammation. However, adenosine A2ARc signaling can sociated with reduced TNF-a bioactivity because of high
attenuate DC activation and protect from injury in ischemic serum levels of TNF receptors, which also correlated with
AKI (56). increased disease activity (67). Finally, in patients with lupus
nephritis, 10 weeks of infliximab treatment reduced protein-
Autoimmune Crescentic Glomerulonephritis uria but increased anti-DNA antibodies. Longer treatment
Lupus Nephritis was associated with adverse effects (68). In patients with
SLE is a disease with evidence of genetic, epigenetic, and RA treated with anti–TNF-a mAbs, lupus syndromes devel-
environmental contributions. There appears to be many oped, anti-DNAs were induced (69), and some patients
different immune abnormalities associated with this dis- developed GN (70).
ease, including significant abnormalities of cytokine circuits. IL-6. IL-6 is likely to act in concert with type 1 IFNs to
It is also likely that there are multiple paths of autoimmunity induce B-cell autoimmunity in SLE. Its levels are elevated
depending in part on which component of immunomodu- in lupus nephritis and correlate with disease activity (71).
lation is defective. Although it is likely that cytokines are IL-6 has been demonstrated in glomerular immune com-
therapeutic targets in this disease, the known presence of plexes and proximal tubular epithelial cells in lupus ne-
multiple immunoregulatory abnormalities suggests that SLE phritis (72). Intrinsic renal cells produce IL-6, and this can
should not be considered as a single homogenous disease. be enhanced by anti-DNA antibodies (73). In lupus-prone
IFN-a. There is good evidence that IFN-a is an impor- mice, IL-6 exacerbates GN while inhibiting IL-6 signaling
tant inducer of antichromatin autoimmunity in patients attenuated GN and reducing autoimmunity, therefore en-
developing SLE. Early in the disease, it has been demon- hancing survival (74). Most IL-6 inhibition trials have oc-
strated that immune complexes (containing DNA and/or curred in RA while data are emerging in SLE. Tocilizumab
RNA) are taken up by plasmacytoid DCs by FCgR-mediated reduced acute phase reactants, anti–double-stranded DNA
internalization. Together with TLR7 and TLR9 stimulation, antibody, and SELENA-SLEDAI scores in patients with
this induces IFN-a production, driving autoimmunity. moderately active lupus nephritis (75).
Evidence supporting this comes from the high incidence A number of biologic interventions target molecules or
of IFN-a–regulated genes in PBMCs of patients with SLE, immune cells upstream of cytokine production. The beneficial
Clin J Am Soc Nephrol 10: 2243–2254, December, 2015 Cytokine: Names and Numbers, Holdsworth and Gan 2249
effects are likely to result from their effects on cytokine animal model of human crescentic GN. There is considerable
mediation of target organ inflammation. data showing that immune cytokines are critically involved
B-cell Activating Factor. B-cell activating factor is also in inducing nephritogenic autoimmunity and mediating glo-
known as B lymphocyte stimulator and is essential for B-cell merular injury in these models. Moreover, studies in this
maturation, survival, and Ig class switching. Its levels are model provide proof of concept that the inhibition of selected
elevated in SLE and correlate with disease activity and flares cytokines can prevent and treat disease.
(76). Belimumab, a humanized anti-B lymphocyte stimulator Numerous innate cytokines have been associated with
mAb has been shown in two trials to demonstrate a modest pathogenesis of anti-GBM GN. A pathogenic role in anti-
but significant benefit in reducing disease activity. Patients GBM GN has been demonstrated for each of the following
with lupus nephritis were excluded, but in one trial, 15% of innate cytokines: GM-CSF, G-CSF, IL-1b, TNF-a, and
patients had evidence of nephritis. A post hoc analysis CXCL8, by studies in cytokine gene–deleted mice. All of
showed significant reduction in proteinuria. Trials in lupus these innate cytokines recruit inflammatory cells to the
nephritis are underway (77,78). kidney and direct the subsequent development of anti-
Abatacept. The data at hand do not provide evidence for GBM GN. Gene deletion of the key Th1 cytokines (IL-12
CTLA4-Ig efficacy in the treatment of lupus nephritis. A and IFN-g) resulted in attenuated crescentic GN, and gene
phase IIb trial involving patients with lupus with poly- deletion of the key Th1 transcription factor (T-bet) is pro-
arthritis and discoid lupus did not meet its primary or tective (85,86). After the discovery of the CD41 Th17 subset,
secondary end point, flare prevention, and the infection studies using mice deficient in p19, p35, and p40 (compo-
incidence was significantly higher in the abatacept arm nents of the key Th1 and Th17 cytokines, IL-12 and IL-23,
(79). In another 12-month trial of abatacept or placebo, intra- respectively) were used to analyze the relative contributions
venous infusion plus steroid and mycophenolate mofetil of each Th subset in anti-GBM GN. Paust et al. demonstrated
were compared in patients with class III and class IV lupus that Th17 cells contributes to anti-GBM GN with the use of
nephritis. Complete response and renal improvement crite- IL-23p192/2 and IL-17A2/2 mice (87), whereas Odobasic
ria were the same in all groups. Infection was not higher in et al. examined the reciprocal relationship between Th1 and
the abatacept group (80). Th17 and demonstrated that early nephritogenic responses
Anti–TNF-Related Weak Inducer of Apoptosis. There were mediated by Th17 cells, but late disease is Th1 depen-
is growing evidence for the anti–TNF-related weak inducer dent. They also demonstrated that each Th subset counter-
of apoptosis (Tweak)/factor inducible 14 pathway in en- regulated the other (88). Furthermore, Steinmetz et al. used
hancing injury in lupus nephritis. In lupus nephritis, mice with gene deletion of RORg-t, the key Th17 transcrip-
Tweak and its receptor are upregulated in renal tubular tion factor, to confirm the participation of the CD41 Th17
cells, inducing proinflammatory cytokines, including IL-6, subset (89). Direct comparison has been made between the
adhesion molecules, and chemokines (81). Immunoneu- effects of transferring Th17 and Th1 polarized ovalbumin
tralization of Tweak decreases renal inflammation in mu- (OVA) specific CD41 TCR transgenic cells into naïve mice
rine models (82), and these mAb are being studied in lupus with OVA planted on the GBM using a non-nephritogenic
nephritis (ClinicalTrials.gov identifier: NCT0130890). anti-GBM/OVA conjugated antibody. Transfer of both CD41
Laquinimod. Laquinimod is a small molecule that im- T-cell clones induced GN. However, transfer of Th1-polarized
munomodulates APCs to redirect Th subset differentia- cells induced a monocyte and macrophage predominant in-
tion with downregulation of IL-6, IL-12, IL-23, IL-17, and filtrate lesion, whereas Th17-polarized cells induced less in-
TNF-a and increased IL-10. In experimental murine lupus, jury with a neutrophil predominant infiltrate (90). To assess
laquinimod delayed the onset of lupus nephritis. When key Th2 cytokines, anti-GBM GN was induced in IL-42/2
administered as a therapeutic, it attenuated disease severity and IL-102/2 mice. Both groups had augmented Th1 re-
by reducing IFN-g and IL-17A production by splenocytes sponses and increased glomerular injury, whereas infusion
while enhancing IL-10 and Treg frequency (83). In a phase of IL-10 attenuated disease. Tregs are not only important in
II study in active lupus nephritis, mycophenolate mofetil maintaining self-tolerance but are also necessary in control-
and high-dose steroid were administered with or without ling overt inflammatory responses. Transfer of Tregs (CD41
laquinimod. Laquinimod had an additive effect with renal CD251) before and after the induction of experimental anti-
function and proteinuria improvement. Adverse effects were GBM GN suppressed the development of GN by reducing
not observed (84). Th1 responses (91). Interestingly, Eller et al. demonstrated
Despite many trials of therapeutics that attenuate cyto- that Treg-derived IL-9 is essential for the recruitment of
kine action being performed in SLE, there is still much that MCs, and both are required to attenuate anti-GBM GN (92).
needs to be understood about the role of cytokines in this Cytokine Production by Resident Renal Cells. Resident
disease. Unfortunately, the simple application of therapies cells within the kidney, including tubular and glomerular
successful in other immune inflammatory diseases (as in cells, interact with infiltrating leukocytes resulting in their
the case of anti–TNF-a immune neutralization) appears to synthesis of injurious TNF-a in response to leukocyte-
be much more problematic in SLE. Finally, the diversity of produced IL-1 (73). The relative roles of leukocyte and
patterns of disease and the involvement of different organs resident cytokine production have been studied in anti-
means lupus nephritis is unfortunately an exclusion in GBM GN in mice using cytokine chimeric mice where the
many trials, denying these patients the opportunity for cytokine gene has been deleted from either bone marrow–
potentially more effective treatments. derived leukocytes or from resident renal cells (86). Nonchi-
Experimental Antiglomerular Basement Membrane meric TNF-a2/2 mice are significantly protected from the
Glomerulonephritis. Experimental antiglomerular base- development of GN. When the TNF-a gene is knocked out
ment membrane (GBM) GN is the most widely studied from resident cells, similar attenuation was observed,
2250 Clinical Journal of the American Society of Nephrology
whereas knockout of the TNF-a gene in bone marrow only target autoantigens, proteinase 3 or myeloperoxidase
caused mild protection, suggesting that TNF-a produced by (MPO) (93). In treatment of refractory disease with T
resident cells is the major source of injurious TNF-a in this cell–specific targeted therapies, antithymocyte globulin
disease. These studies also produced evidence of complex was beneficial and capable of inducing remission (94). Fur-
cytokine interactions between resident cells and infiltrating thermore, cytokine profiling of biopsied nasal mucosal tis-
leukocytes. Studies with IL-1b2/2 and IL-1R12/2 mice sue, bronchoalveolar lavage, and PBMCs from patients
showed that IL-1b mainly derived from leukocytes actives with granulomatous polyangiitis demonstrated increased
IL-1R1 on resident cells, inducing TNF-a production that expression of IFN-g, denoting a Th1 cytokine pattern (95).
causes significant glomerular injury (86) (Figure 2). Nogueira et al. found that in the serum of acute or convales-
Autoimmune Anti-GBM Glomerulonephritis. In auto- cent AAV patients, IL-17A and IL-23 levels were increased,
immune anti-GBM GN, the target autoantigen is the non- and this correlated with disease severity and ANCA titer
collagenase domain of the alpha 3 chain of type IV collagen. (96). IL-6 was also elevated in active disease. Chavele et al.
Immunization with this antigen induces autoimmunity shown reported that in patients with MPO-AAV, MPO-stimulated
by the production of circulating anti-GBM antibodies and the recall responses showed elevated IFN-g (97). Collectively,
development of crescentic GN. IFN-g2/2 mice developed these data provide evidence in support of both Th1 and
worse disease. The relative contributions of Th1 and Th17 Th17 involvement in AAV. Furthermore, IL-17–producing
to disease development were assessed using p352/2 (defi- cells were found in renal biopsies from patients with acute
cient in Th1 subset) and p192/2 (deficient in Th17 subset) vasculitis, and most of the IL-17–positive cells present were
mice. The p192/2 mice were protected from GN, but the innate leukocytes (neutrophils and MCs) (98).
p352/2 mice were unaffected compared with controls, In experimental MPO-ANCA GN, IL-17A2/2 mice
confirming a pathogenic role for Th17 but not for Th1 in were protected from the development of anti-MPO auto-
this disease. immunity and glomerular injury (99). Immunoneutraliza-
ANCA-Associated Crescentic Glomerulonephritis. The tion of TNF-a caused significant reduction in lung
most common cause of crescentic GN is ANCA-associated hemorrhage and renal injury in Wistar–Kyoto rats with
vasculitis (AAV). Evidence suggests cytokines directing induced anti-MPO AAV (100).
the underlying nephritogenic autoimmunity are likely to The only cytokine-based clinical trials in AAV have been
be therapeutic targets that could be neutralized with avail- of anti–TNF-a. Treatment with etanercept (anti–TNF-a
able biologic agents. mAb) was not effective and was associated with a high
PBMCs from patients with ANCA-associated GN have rate of treatment-related adverse side effects (101). A
CD41 T cells that proliferate when stimulated with the smaller clinical trial using adalimumab with prednisolone
Figure 2. | Innate macrophages produce IL-1 which binds to IL-1Rc on intrinsic renal cells (endothelial cells, podocytes, and mesangial cells)
to produce injurious TNF-a that amplifies T effector cell responses resulting in crescent formation and glomerular injury. Rc, receptor.
Clin J Am Soc Nephrol 10: 2243–2254, December, 2015 Cytokine: Names and Numbers, Holdsworth and Gan 2251
plus cyclophosphamide showed similar efficacy to using 6. Crotty S: Follicular helper CD4 T cells (TFH). Annu Rev Im-
these drugs but afforded with less steroid exposure (102). munol 29: 621–663, 2011
7. Auphan N, DiDonato JA, Rosette C, Helmberg A, Karin M:
Immunosuppression by glucocorticoids: Inhibition of NF-kappa
Opportunities for Introducing Biologic Therapies to Treat B activity through induction of I kappa B synthesis. Science 270:
286–290, 1995
Renal Diseases 8. Göttlicher M, Heck S, Herrlich P: Transcriptional cross-talk, the
The evidence outlined here suggests that there are second mode of steroid hormone receptor action. J Mol Med
important renal diseases that may benefit from the appli- (Berl) 76: 480–489, 1998
cation of well targeted biologic therapies on the basis of 9. Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy
cytokine manipulation. KM: Th17: An effector CD4 T cell lineage with regulatory T cell
ties. Immunity 24: 677–688, 2006
Innate cytokines are prominent participants in many 10. Kiely PD, Johnson D, Bourke BE: An open study of oxpentifylline
forms of AKI. Moreover, in some clinical settings, such as in early rheumatoid arthritis. Br J Rheumatol 37: 1033–1035,
renal transplantation, high-risk surgery (e.g., elective cor- 1998
onary grafting), and for optimal use of effective chemo- 11. Elliott MJ, Maini RN, Feldmann M, Kalden JR, Antoni C, Smolen
therapeutics (nephrotoxic dose-limited like cisplatin), the JS, Leeb B, Breedveld FC, Macfarlane JD, Bijl H: Randomised
double-blind comparison of chimeric monoclonal antibody to
opportunity exists to preemptively block likely injurious tumour necrosis factor alpha (cA2) versus placebo in rheuma-
cytokine pathways.There is a need for more selective, less toid arthritis. Lancet 344: 1105–1110, 1994
toxic therapies to treat autoimmune inflammatory forms of 12. Braun J, Brandt J, Listing J, Zink A, Alten R, Golder W,
proliferative/crescentic GN, including lupus nephritis, Gromnica-Ihle E, Kellner H, Krause A, Schneider M, Sörensen
H, Zeidler H, Thriene W, Sieper J: Treatment of active anky-
anti-GBM, and ANCA-associated disease. These represent
losing spondylitis with infliximab: A randomised controlled
the more serious and inflammatory categories of theses multicentre trial. Lancet 359: 1187–1193, 2002
autoimmune diseases. They share features of other diseases, 13. Rutgeerts P, Sandborn WJ, Feagan BG, Reinisch W, Olson A,
such as RA and IBD, where new biologics are now part of Johanns J, Travers S, Rachmilewitz D, Hanauer SB, Lichtenstein
the therapeutic pharmacopeia. GR, de Villiers WJ, Present D, Sands BE, Colombel JF: Infliximab
for induction and maintenance therapy for ulcerative colitis. N
The initial attempt to introduce anti–TNF-a mAbs in Engl J Med 353: 2462–2476, 2005
AAV was disappointing. It should remind us that trans- 14. Stone JH, Holbrook JT, Marriott MA, Tibbs AK, Sejismundo LP,
lation of therapies from one disease to another is not sim- Min YI, Specks U, Merkel PA, Spiera R, Davis JC, St Clair EW,
ple or without risk, but without well managed risk, there McCune WJ, Ytterberg SR, Allen NB, Hoffman GS; Wegener’s
will be no progress. Perhaps we should apply the princi- Granulomatosis Etanercept Trial Research Group: Solid malig-
nancies among patients in the Wegener’s Granulomatosis
ples behind the introduction of anti–TNF-a to RA, use tis- Etanercept Trial. Arthritis Rheum 54: 1608–1618, 2006
sue samples from patients with active disease to assess the 15. The Lenercept Multiple Sclerosis Study Group and The Uni-
dominant cytokines in active untreated disease, assess the versity of British Columbia MS/MRI Analysis Group: TNF neu-
effects of these cytokines on relevant renal tissues in vitro, tralization in MS: Results of a randomized, placebo-controlled
and use relevant animal models to provide proof of con- multicenter study. Neurology 53: 457–465, 1999
16. Probert L, Eugster HP, Akassoglou K, Bauer J, Frei K, Lassmann
cept for the specificity, efficacy, and minimal toxicity in H, Fontana A: TNFR1 signalling is critical for the development
preclinical trials. Clinical trials with the greatest likelihood of demyelination and the limitation of T-cell responses during
of success are those that target dominant cytokines in renal immune-mediated CNS disease. Brain 123: 2005–2019, 2000
diseases using immunoneutralising mAbs where minimal 17. Feldmann M, Maini RN: Anti-TNF therapy, from rationale to
standard of care: What lessons has it taught us? J Immunol 185:
toxicity and clinical effectiveness has already been shown
791–794, 2010
in other chronic autoimmune and or autoinflammatory 18. Smolen JS, Beaulieu A, Rubbert-Roth A, Ramos-Remus C,
disease. Using these criteria we could now be planning Rovensky J, Alecock E, Woodworth T, Alten R; OPTION In-
clinical trials for targeting the major innate cytokine as vestigators: Effect of interleukin-6 receptor inhibition with
prophylaxis and treatment and considering neutralizing tocilizumab in patients with rheumatoid arthritis (OPTION
study): A double-blind, placebo-controlled, randomised trial.
mAbs to IL-17 and IL-23 in ANCA vasculitis. Lancet 371: 987–997, 2008
19. De Benedetti F, Brunner HI, Ruperto N, Kenwright A, Wright S,
Disclosures Calvo I, Cuttica R, Ravelli A, Schneider R, Woo P, Wouters C,
None. Xavier R, Zemel L, Baildam E, Burgos-Vargas R, Dolezalova P,
Garay SM, Merino R, Joos R, Grom A, Wulffraat N, Zuber Z,
Zulian F, Lovell D, Martini A; PRINTO; PRCSG: Randomized
References trial of tocilizumab in systemic juvenile idiopathic arthritis. N
1. Eleftheriadis T, Pissas G, Liakopoulos V, Stefanidis I, Lawson BR: Engl J Med 367: 2385–2395, 2012
Toll-like receptors and their role in renal pathologies. Inflamm 20. Dinarello CA, Simon A, van der Meer JW: Treating inflammation
by blocking interleukin-1 in a broad spectrum of diseases. Nat
Allergy Drug Targets 11: 464–477, 2012
Rev Drug Discov 11: 633–652, 2012
2. Gabay C, Kushner I: Acute-phase proteins and other systemic
21. Webster AC, Ruster LP, McGee R, Matheson SL, Higgins GY,
responses to inflammation. N Engl J Med 340: 448–454, 1999
Willis NS, Chapman JR, Craig JC: Interleukin 2 receptor an-
3. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman
tagonists for kidney transplant recipients. Cochrane Database
RL: Two types of murine helper T cell clone. I. Definition ac- Syst Rev (1): CD003897, 2010
cording to profiles of lymphokine activities and secreted pro- 22. Nussenblatt RB, Fortin E, Schiffman R, Rizzo L, Smith J, Van
teins. J Immunol 136: 2348–2357, 1986 Veldhuisen P, Sran P, Yaffe A, Goldman CK, Waldmann TA,
4. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Whitcup SM: Treatment of noninfectious intermediate and
Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ: IL-23 posterior uveitis with the humanized anti-Tac mAb: A phase I/II
drives a pathogenic T cell population that induces autoimmune clinical trial. Proc Natl Acad Sci U S A 96: 7462–7466, 1999
inflammation. J Exp Med 201: 233–240, 2005 23. Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR,
5. Kaplan MH: Th9 cells: Differentiation and disease. Immunol Katz S, Johanns J, Blank M, Rutgeerts P; Ustekinumab Crohn’s
Rev 252: 104–115, 2013 Disease Study Group: A randomized trial of Ustekinumab, a
2252 Clinical Journal of the American Society of Nephrology
human interleukin-12/23 monoclonal antibody, in patients with 38. Mun SH, Kim JW, Nah SS, Ko NY, Lee JH, Kim JD, Kim K, Kim
moderate-to-severe Crohn’s disease. Gastroenterology 135: HS, Choi JD, Kim SH, Lee CK, Park SH, Kim BK, Kim HS, Kim
1130–1141, 2008 YM, Choi WS: Tumor necrosis factor alpha-induced interleukin-
24. McInnes IB, Kavanaugh A, Gottlieb AB, Puig L, Rahman P, 32 is positively regulated via the Syk/protein kinase Cdelta/JNK
Ritchlin C, Brodmerkel C, Li S, Wang Y, Mendelsohn AM, Doyle pathway in rheumatoid synovial fibroblasts. Arthritis Rheum 60:
MK; PSUMMIT 1 Study Group: Efficacy and safety of ustekinumab 678–685, 2009
in patients with active psoriatic arthritis: 1 year results of the 39. Vallés PG, Lorenzo AG, Bocanegra V, Vallés R: Acute kidney
phase 3, multicentre, double-blind, placebo-controlled injury: what part do toll-like receptors play? Int J Nephrol
PSUMMIT 1 trial. Lancet 382: 780–789, 2013 Renovasc Dis 7: 241–251, 2014
25. Leonardi C, Matheson R, Zachariae C, Cameron G, Li L, Edson- 40. Kinsey GR, Okusa MD: Role of leukocytes in the pathogenesis
Heredia E, Braun D, Banerjee S: Anti-interleukin-17 mono- of acute kidney injury. Crit Care 16: 214, 2012
clonal antibody ixekizumab in chronic plaque psoriasis. N Engl 41. Summers SA, Chan J, Gan PY, Dewage L, Nozaki Y, Steinmetz
J Med 366: 1190–1199, 2012 OM, Nikolic-Paterson DJ, Kitching AR, Holdsworth SR: Mast
26. Genovese MC, Durez P, Richards HB, Supronik J, Dokoupilova cells mediate acute kidney injury through the production of
E, Mazurov V, Aelion JA, Lee SH, Codding CE, Kellner H, Ikawa TNF. J Am Soc Nephrol 22: 2226–2236, 2011
T, Hugot S, Mpofu S: Efficacy and safety of secukinumab in 42. Donnahoo KK, Meng X, Ayala A, Cain MP, Harken AH,
patients with rheumatoid arthritis: A phase II, dose-finding, Meldrum DR: Early kidney TNF-alpha expression mediates
double-blind, randomised, placebo controlled study. Ann neutrophil infiltration and injury after renal ischemia-
Rheum Dis 72: 863–869, 2013 reperfusion. Am J Physiol 277: R922–R929, 1999
27. Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, 43. Akcay A, Nguyen Q, Edelstein CL: Mediators of inflammation in
Reinisch W, Higgins PD, Wehkamp J, Feagan BG, Yao MD, acute kidney injury. Mediators Inflamm 2009: 137072, 2009
Karczewski M, Karczewski J, Pezous N, Bek S, Bruin G, 44. Burne MJ, Elghandour A, Haq M, Saba SR, Norman J, Condon T,
Mellgard B, Berger C, Londei M, Bertolino AP, Tougas G, Travis Bennett F, Rabb H: IL-1 and TNF independent pathways medi-
SP; Secukinumab in Crohn’s Disease Study Group: ate ICAM-1/VCAM-1 up-regulation in ischemia reperfusion
Secukinumab, a human anti-IL-17A monoclonal antibody, for injury. J Leukoc Biol 70: 192–198, 2001
moderate to severe Crohn’s disease: Unexpected results of a 45. Kim HJ, Lee DW, Ravichandran K, O Keys D, Akcay A, Nguyen
randomised, double-blind placebo-controlled trial. Gut 61: Q, He Z, Jani A, Ljubanovic D, Edelstein CL: NLRP3 in-
1693–1700, 2012 flammasome knockout mice are protected against ischemic but
28. A Phase II, Multicenter, Randomized, Double-Blind, Parallel not cisplatin-induced acute kidney injury. J Pharmacol Exp Ther
Group, Placebo-Controlled, Adaptive Dose-Ranging Study to 346: 465–472, 2013
Evaluate the Efficacy and Safety of AIN457 (Secukinumab) in 46. Faubel S, Ljubanovic D, Reznikov L, Somerset H, Dinarello CA,
Patients with Relapsing Multiple Sclerosis. Available at: https:// Edelstein CL: Caspase-1-deficient mice are protected against
clinicaltrials.gov/show/NCT01874340. Accessed August 1, 2014 cisplatin-induced apoptosis and acute tubular necrosis. Kidney
29. Maini RN, Paulus H, Breedveld FC, Moreland LW, William E, Int 66: 2202–2213, 2004
Russell AS, Charles P, Davies D, Grint P, Wherry JC, Feldmann 47. Faubel S, Lewis EC, Reznikov L, Ljubanovic D, Hoke TS,
M: rHuIL-10 in subjects with active rheumatoid arthritis (RA): Somerset H, Oh DJ, Lu L, Klein CL, Dinarello CA, Edelstein CL:
A phase I and cytokine response study. Arthritis Rheum 40: Cisplatin-induced acute renal failure is associated with an in-
S224, 1997 crease in the cytokines interleukin (IL)-1beta, IL-18, IL-6, and
30. McInnes IB, Illei GG, Danning CL, Yarboro CH, Crane M, neutrophil infiltration in the kidney. J Pharmacol Exp Ther 322:
Kuroiwa T, Schlimgen R, Lee E, Foster B, Flemming D, Prussin C, 8–15, 2007
Fleisher TA, Boumpas DT: IL-10 improves skin disease and 48. Patel NS, Chatterjee PK, Di Paola R, Mazzon E, Britti D, De Sarro
modulates endothelial activation and leukocyte effector func- A, Cuzzocrea S, Thiemermann C: Endogenous interleukin-6
tion in patients with psoriatic arthritis. J Immunol 167: 4075– enhances the renal injury, dysfunction, and inflammation
4082, 2001 caused by ischemia/reperfusion. J Pharmacol Exp Ther 312:
31. Fedorak RN, Gangl A, Elson CO, Rutgeerts P, Schreiber S, Wild 1170–1178, 2005
G, Hanauer SB, Kilian A, Cohard M, LeBeaut A, Feagan B: Re- 49. Nechemia-Arbely Y, Barkan D, Pizov G, Shriki A, Rose-John S,
combinant human interleukin 10 in the treatment of patients Galun E, Axelrod JH: IL-6/IL-6R axis plays a critical role in acute
with mild to moderately active Crohn’s disease. The Interleukin kidney injury. J Am Soc Nephrol 19: 1106–1115, 2008
10 Inflammatory Bowel Disease Cooperative Study Group. 50. Deng J, Kohda Y, Chiao H, Wang Y, Hu X, Hewitt SM, Miyaji T,
Gastroenterology 119: 1473–1482, 2000 McLeroy P, Nibhanupudy B, Li S, Star RA: Interleukin-10 in-
32. Maxwell LJ, Singh JA: Abatacept for rheumatoid arthritis: A hibits ischemic and cisplatin-induced acute renal injury. Kidney
Cochrane systematic review. J Rheumatol 37: 234–245, 2010 Int 60: 2118–2128, 2001
33. Evans CH, Robbins PD, Ghivizzani SC, Wasko MC, Tomaino 51. Lee H, Nho D, Chung HS, Lee H, Shin MK, Kim SH, Bae H:
MM, Kang R, Muzzonigro TA, Vogt M, Elder EM, Whiteside TL, CD41CD251 regulatory T cells attenuate cisplatin-induced
Watkins SC, Herndon JH: Gene transfer to human joints: nephrotoxicity in mice. Kidney Int 78: 1100–1109, 2010
Progress toward a gene therapy of arthritis. Proc Natl Acad Sci U 52. Kinsey GR, Sharma R, Huang L, Li L, Vergis AL, Ye H, Ju ST,
S A 102: 8698–8703, 2005 Okusa MD: Regulatory T cells suppress innate immunity in
34. Sengupta TK, Schmitt EM, Ivashkiv LB: Inhibition of cytokines kidney ischemia-reperfusion injury. J Am Soc Nephrol 20:
and JAK-STAT activation by distinct signaling pathways. Proc 1744–1753, 2009
Natl Acad Sci U S A 93: 9499–9504, 1996 53. Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Satpute
35. van Vollenhoven RF, Fleischmann R, Cohen S, Lee EB, Garcı́a SR, Crow MT, King LS, Rabb H: Foxp31 regulatory T cells
Meijide JA, Wagner S, Forejtova S, Zwillich SH, Gruben D, participate in repair of ischemic acute kidney injury. Kidney Int
Koncz T, Wallenstein GV, Krishnaswami S, Bradley JD, 76: 717–729, 2009
Wilkinson B; ORAL Standard Investigators: Tofacitinib or 54. Ratliff BB, Rabadi MM, Vasko R, Yasuda K, Goligorsky MS:
adalimumab versus placebo in rheumatoid arthritis. N Engl J Messengers without borders: Mediators of systemic in-
Med 367: 508–519, 2012 flammatory response in AKI. J Am Soc Nephrol 24: 529–536,
36. Pine PR, Chang B, Schoettler N, Banquerigo ML, Wang S, Lau A, 2013
Zhao F, Grossbard EB, Payan DG, Brahn E: Inflammation and 55. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M,
bone erosion are suppressed in models of rheumatoid arthritis Che J, Frazier A, Yang H, Ivanova S, Borovikova L, Manogue KR,
following treatment with a novel Syk inhibitor. Clin Immunol Faist E, Abraham E, Andersson J, Andersson U, Molina PE,
124: 244–257, 2007 Abumrad NN, Sama A, Tracey KJ: HMG-1 as a late mediator of
37. Weinblatt ME, Kavanaugh A, Genovese MC, Musser TK, endotoxin lethality in mice. Science 285: 248–251, 1999
Grossbard EB, Magilavy DB: An oral spleen tyrosine kinase 56. Okusa MD, Li L: Dendritic cells in acute kidney injury: cues
(Syk) inhibitor for rheumatoid arthritis. N Engl J Med 363: 1303– from the microenvironment. Trans Am Clin Climatol Assoc 123:
1312, 2010 54–62, discussion 62–63, 2012
Clin J Am Soc Nephrol 10: 2243–2254, December, 2015 Cytokine: Names and Numbers, Holdsworth and Gan 2253
57. Rönnblom L, Eloranta ML, Alm GV: The type I interferon system 73. Yung S, Cheung KF, Zhang Q, Chan TM: Mediators of
in systemic lupus erythematosus. Arthritis Rheum 54: 408–420, inflammation and their effect on resident renal cells: im-
2006 plications in lupus nephritis. Clin Dev Immunol 2013: 317682,
58. Feng X, Wu H, Grossman JM, Hanvivadhanakul P, FitzGerald 2013
JD, Park GS, Dong X, Chen W, Kim MH, Weng HH, Furst DE, 74. Cash H, Relle M, Menke J, Brochhausen C, Jones SA, Topley N,
Gorn A, McMahon M, Taylor M, Brahn E, Hahn BH, Tsao BP: Galle PR, Schwarting A: Interleukin 6 (IL-6) deficiency delays
Association of increased interferon-inducible gene expression lupus nephritis in MRL-Faslpr mice: The IL-6 pathway as a
with disease activity and lupus nephritis in patients with new therapeutic target in treatment of autoimmune kidney
systemic lupus erythematosus. Arthritis Rheum 54: 2951–2962, disease in systemic lupus erythematosus. J Rheumatol 37: 60–
2006 70, 2010
59. Criswell LA: The genetic contribution to systemic lupus eryth- 75. Illei GG, Shirota Y, Yarboro CH, Daruwalla J, Tackey E, Takada
ematosus. Bull NYU Hosp Jt Dis 66: 176–183, 2008 K, Fleisher T, Balow JE, Lipsky PE: Tocilizumab in systemic
60. Petri M, Wallace DJ, Spindler A, Chindalore V, Kalunian K, lupus erythematosus: Data on safety, preliminary efficacy,
Mysler E, Neuwelt CM, Robbie G, White WI, Higgs BW, Yao Y, and impact on circulating plasma cells from an open-label
Wang L, Ethgen D, Greth W: Sifalimumab, a human anti- phase I dosage-escalation study. Arthritis Rheum 62: 542–552,
interferon-a monoclonal antibody, in systemic lupus eryth- 2010
ematosus: A phase I randomized, controlled, dose-escalation 76. Petri M, Stohl W, Chatham W, McCune WJ, Chevrier M, Ryel J,
study. Arthritis Rheum 65: 1011–1021, 2013 Recta V, Zhong J, Freimuth W: Association of plasma B
61. Tcherepanova I, Curtis M, Sale M, Miesowicz F, Nicolette C: lymphocyte stimulator levels and disease activity in systemic
Results of a randomized placebo controlled phase IA study of lupus erythematosus. Arthritis Rheum 58: 2453–2459,
AGS-009, a humanized anti-interferon-alpha monoclonal an- 2008
tibody in subjects with systemic lupus erythematosus. Ann 77. Navarra SV, Guzmán RM, Gallacher AE, Hall S, Levy RA,
Rheum Dis 71[Suppl 3]: 536–537, 2013 Jimenez RE, Li EK, Thomas M, Kim HY, León MG, Tanasescu C,
62. Lauwerys BR, Hachulla E, Spertini F, Lazaro E, Jorgensen C, Nasonov E, Lan JL, Pineda L, Zhong ZJ, Freimuth W, Petri MA;
Mariette X, Haelterman E, Grouard-Vogel G, Fanget B, Dhellin BLISS-52 Study Group: Efficacy and safety of belimumab in
O, Vandepapelière P, Houssiau FA: Down-regulation of in- patients with active systemic lupus erythematosus: A random-
terferon signature in systemic lupus erythematosus patients by ised, placebo-controlled, phase 3 trial. Lancet 377: 721–731,
active immunization with interferon a-kinoid. Arthritis Rheum 2011
65: 447–456, 2013 78. Furie R, Petri M, Zamani O, Cervera R, Wallace DJ, Tegzová D,
63. Brennan DC, Yui MA, Wuthrich RP, Kelley VE: Tumor necrosis Sanchez-Guerrero J, Schwarting A, Merrill JT, Chatham WW,
factor and IL-1 in New Zealand Black/White mice. Enhanced Stohl W, Ginzler EM, Hough DR, Zhong ZJ, Freimuth W, van
gene expression and acceleration of renal injury. J Immunol Vollenhoven RF; BLISS-76 Study Group: A phase III, random-
ized, placebo-controlled study of belimumab, a monoclonal
143: 3470–3475, 1989
64. Bethunaickan R, Sahu R, Liu Z, Tang YT, Huang W, Edegbe O, antibody that inhibits B lymphocyte stimulator, in patients with
systemic lupus erythematosus. Arthritis Rheum 63: 3918–3930,
Tao H, Ramanujam M, Madaio MP, Davidson A: Anti-tumor
2011
necrosis factor a treatment of interferon-a-induced murine lu-
79. Merrill JT, Burgos-Vargas R, Westhovens R, Chalmers A, D’Cruz
pus nephritis reduces the renal macrophage response but does
D, Wallace DJ, Bae SC, Sigal L, Becker JC, Kelly S, Raghupathi
not alter glomerular immune complex formation. Arthritis
K, Li T, Peng Y, Kinaszczuk M, Nash P: The efficacy and safety
Rheum 64: 3399–3408, 2012
of abatacept in patients with non-life-threatening manifes-
65. Gigante A, Gasperini ML, Afeltra A, Barbano B, Margiotta D,
tations of systemic lupus erythematosus: Results of a twelve-
Cianci R, De Francesco I, Amoroso A: Cytokines expression in
month, multicenter, exploratory, phase IIb, randomized,
SLE nephritis. Eur Rev Med Pharmacol Sci 15: 15–24, 2011
double-blind, placebo-controlled trial. Arthritis Rheum 62:
66. Yu CL, Chang KL, Chiu CC, Chiang BN, Han SH, Wang SR:
3077–3087, 2010
Defective phagocytosis, decreased tumour necrosis factor- 80. Wofsy D, Hillson JL, Diamond B: Abatacept for lupus nephritis:
alpha production, and lymphocyte hyporesponsiveness Alternative definitions of complete response support conflicting
predispose patients with systemic lupus erythematosus to conclusions. Arthritis Rheum 64: 3660–3665, 2012
infections. Scand J Rheumatol 18: 97–105, 1989 81. Sanz AB, Justo P, Sanchez-Ni~ no MD, Blanco-Colio LM, Winkles
67. Aderka D, Wysenbeek A, Engelmann H, Cope AP, Brennan F, JA, Kreztler M, Jakubowski A, Blanco J, Egido J, Ruiz-Ortega M,
Molad Y, Hornik V, Levo Y, Maini RN, Feldmann M, Wallach D: Ortiz A: The cytokine TWEAK modulates renal
Correlation between serum levels of soluble tumor necrosis tubulointerstitial inflammation. J Am Soc Nephrol 19: 695–703,
factor receptor and disease activity in systemic lupus eryth- 2008
ematosus. Arthritis Rheum 36: 1111–1120, 1993 82. Xia Y, Campbell SR, Broder A, Herlitz L, Abadi M, Wu P,
68. Aringer M, Houssiau F, Gordon C, Graninger WB, Voll RE, Rath Michaelson JS, Burkly LC, Putterman C: Inhibition of the
E, Steiner G, Smolen JS: Adverse events and efficacy of TNF- TWEAK/Fn14 pathway attenuates renal disease in nephrotoxic
alpha blockade with infliximab in patients with systemic lupus serum nephritis. Clin Immunol 145: 108–121, 2012
erythematosus: Long-term follow-up of 13 patients. Rheuma- 83. Lourenço EV, Wong M, Hahn BH, Palma-Diaz MF, Skaggs BJ:
tology (Oxford) 48: 1451–1454, 2009 Laquinimod delays and suppresses nephritis in lupus-prone
69. Charles PJ, Smeenk RJ, De Jong J, Feldmann M, Maini RN: As- mice and affects both myeloid and lymphoid immune cells.
sessment of antibodies to double-stranded DNA induced in Arthritis Rheum (Munch) 66: 674–685, 2014
rheumatoid arthritis patients following treatment with 84. Jayne D, Appel G, Chan TM, Barkay H, Weiss R, Wofsy D: A
infliximab, a monoclonal antibody to tumor necrosis factor al- randomized controlled study of laquinimod in active lupus
pha: Findings in open-label and randomized placebo- nephritis patients in combination with standard care. Ann
controlled trials. Arthritis Rheum 43: 2383–2390, 2000 Rheum Dis 72[Suppl 3]: A164, 2013
70. Stokes MB, Foster K, Markowitz GS, Ebrahimi F, Hines W, 85. Phoon RK, Kitching AR, Odobasic D, Jones LK, Semple TJ,
Kaufman D, Moore B, Wolde D, D’Agati VD: Development of Holdsworth SR: T-bet deficiency attenuates renal injury in ex-
glomerulonephritis during anti-TNF-alpha therapy for rheu- perimental crescentic glomerulonephritis. J Am Soc Nephrol
matoid arthritis. Nephrol Dial Transplant 20: 1400–1406, 2005 19: 477–485, 2008
71. Chun HY, Chung JW, Kim HA, Yun JM, Jeon JY, Ye YM, Kim SH, 86. Tipping PG, Holdsworth SR: Cytokines in glomerulonephritis.
Park HS, Suh CH: Cytokine IL-6 and IL-10 as biomarkers in Semin Nephrol 27: 275–285, 2007
systemic lupus erythematosus. J Clin Immunol 27: 461–466, 87. Paust HJ, Turner JE, Steinmetz OM, Peters A, Heymann F,
2007 Hölscher C, Wolf G, Kurts C, Mittrücker HW, Stahl RA, Panzer
72. Malide D, Russo P, Bendayan M: Presence of tumor necrosis U: The IL-23/Th17 axis contributes to renal injury in experi-
factor alpha and interleukin-6 in renal mesangial cells of lupus mental glomerulonephritis. J Am Soc Nephrol 20: 969–979,
nephritis patients. Hum Pathol 26: 558–564, 1995 2009
2254 Clinical Journal of the American Society of Nephrology
88. Odobasic D, Gan PY, Summers SA, Semple TJ, Muljadi RC, 96. Nogueira E, Hamour S, Sawant D, Henderson S, Mansfield N,
Iwakura Y, Kitching AR, Holdsworth SR: Interleukin-17A pro- Chavele KM, Pusey CD, Salama AD: Serum IL-17 and IL-23
motes early but attenuates established disease in crescentic levels and autoantigen-specific Th17 cells are elevated in pa-
glomerulonephritis in mice. Am J Pathol 179: 1188–1198, 2011 tients with ANCA-associated vasculitis. Nephrol Dial Trans-
89. Steinmetz OM, Summers SA, Gan PY, Semple T, Holdsworth SR, plant 25: 2209–2217, 2010
Kitching AR: The Th17-defining transcription factor RORgt 97. Chavele KM, Shukla D, Keteepe-Arachi T, Seidel JA, Fuchs D,
promotes glomerulonephritis. J Am Soc Nephrol 22: 472–483, Pusey CD, Salama AD: Regulation of myeloperoxidase-
2011 specific T cell responses during disease remission in anti-
90. Summers SA, Steinmetz OM, Li M, Kausman JY, Semple T, neutrophil cytoplasmic antibody-associated vasculitis: The
Edgtton KL, Borza DB, Braley H, Holdsworth SR, Kitching AR: role of Treg cells and tryptophan degradation. Arthritis Rheum
Th1 and Th17 cells induce proliferative glomerulonephritis. J 62: 1539–1548, 2010
Am Soc Nephrol 20: 2518–2524, 2009 98. Velden J, Paust HJ, Hoxha E, Turner JE, Steinmetz OM, Wolf G,
91. Wolf D, Hochegger K, Wolf AM, Rumpold HF, Gastl G, Tilg H, Jabs WJ, Özcan F, Beige J, Heering PJ, Schröder S, Kneißler
Mayer G, Gunsilius E, Rosenkranz AR: CD41CD251 regula- U, Disteldorf E, Mittrücker HW, Stahl RA, Helmchen U,
tory T cells inhibit experimental anti-glomerular basement Panzer U: Renal IL-17 expression in human ANCA-
membrane glomerulonephritis in mice. J Am Soc Nephrol 16: associated glomerulonephritis. Am J Physiol Renal Physiol
302: F1663–F1673, 2012
1360–1370, 2005
99. Gan PY, Steinmetz OM, Tan DS, O’Sullivan KM, Ooi JD,
92. Eller K, Wolf D, Huber JM, Metz M, Mayer G, McKenzie AN,
Iwakura Y, Kitching AR, Holdsworth SR: Th17 cells promote
Maurer M, Rosenkranz AR, Wolf AM: IL-9 production by reg-
autoimmune anti-myeloperoxidase glomerulonephritis. J Am
ulatory T cells recruits mast cells that are essential for regulatory
Soc Nephrol 21: 925–931, 2010
T cell-induced immune suppression. J Immunol 186: 83–91, 100. Little MA, Bhangal G, Smyth CL, Nakada MT, Cook HT,
2011 Nourshargh S, Pusey CD: Therapeutic effect of anti-TNF-alpha
93. King WJ, Brooks CJ, Holder R, Hughes P, Adu D, Savage CO: T antibodies in an experimental model of anti-neutrophil cyto-
lymphocyte responses to anti-neutrophil cytoplasmic plasm antibody-associated systemic vasculitis. J Am Soc
autoantibody (ANCA) antigens are present in patients Nephrol 17: 160–169, 2006
with ANCA-associated systemic vasculitis and persist dur- 101. Wegener’s Granulomatosis Etanercept Trial (WGET) Research
ing disease remission. Clin Exp Immunol 112: 539–546, 1998 Group: Etanercept plus standard therapy for Wegener’s gran-
94. Schmitt WH, Hagen EC, Neumann I, Nowack R, Flores-Suárez ulomatosis. N Engl J Med 352: 351–361, 2005
LF, van der Woude FJ; European Vasculitis Study Group: Treat- 102. Laurino S, Chaudhry A, Booth A, Conte G, Jayne D: Prospective
ment of refractory Wegener’s granulomatosis with antithymo- study of TNFalpha blockade with adalimumab in ANCA-
cyte globulin (ATG): An open study in 15 patients. Kidney Int 65: associated systemic vasculitis with renal involvement. Nephrol
1440–1448, 2004 Dial Transplant 25: 3307–3314, 2010
95. Csernok E, Trabandt A, Müller A, Wang GC, Moosig F, Paulsen J,
Schnabel A, Gross WL: Cytokine profiles in Wegener’s gran-
ulomatosis: Predominance of type 1 (Th1) in the granulomatous Published online ahead of print. Publication date available at www.
inflammation. Arthritis Rheum 42: 742–750, 1999 cjasn.org.