ArticlePDF Available

Inhibitor of NF B Kinase Subunit 2 Blockade Hinders the Initiation but Aggravates the Progression of Crescentic GN

Authors:

Abstract and Figures

The NFκB transcription factor family facilitates the activation of dendritic cells (DCs) and CD4(+) T helper (Th) cells, which are important for protective adaptive immunity. Inappropriate activation of these immune cells may cause inflammatory disease, and NFκB inhibitors are promising anti-inflammatory drug candidates. Here, we investigated whether inhibiting the NFκB-inducing kinase IKK2 can attenuate crescentic GN, a severe DC- and Th cell-dependent kidney inflammatory disease. Prophylactic pharmacologic IKK2 inhibition reduced DC and Th cell activation and ameliorated nephrotoxic serum-induced GN in mice. However, therapeutic IKK2 inhibition during ongoing disease aggravated the nephritogenic immune response and disease symptoms. This effect resulted from the renal loss of regulatory T cells, which have been shown to protect against crescentic GN and which require IKK2. In conclusion, although IKK2 inhibition can suppress the induction of nephritogenic immune responses in vivo, it may aggravate such responses in clinically relevant situations, because it also impairs regulatory T cells and thereby, unleashes preexisting nephritogenic responses. Our findings argue against using IKK2 inhibitors in chronic GN and perhaps, other immune-mediated diseases.
Content may be subject to copyright.
BRIEF COMMUNICATION www.jasn.org
Inhibitor of NFkB Kinase Subunit 2 Blockade Hinders the
Initiation but Aggravates the Progression of Crescentic
GN
Janine Gotot,* Eveline Piotrowski,
Martin S. Otte,*
André P. Tittel,* Guo Linlin,
Chen Yao,
Karl Ziegelbauer,
§
Ulf Panzer,
Natalio Garbi,* Christian Kurts,* and Friedrich Thaiss
*Institute of Experimental Immunology, Rheinische Friedrich Wilhelms University, Bonn, Germany;
Third Medical
Department of Clinical Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany;
Department of
Otorhinolaryngology, Head and Neck Surgery, University of Cologne Germany; and
§
Global Drug Discovery,
TRG Oncology/GT, Bayer Pharma AG, Berlin, Germany
ABSTRACT
The NFkB transcription factor family facilitates the activation of dendritic cells (DCs)
and CD4
+
T helper (Th) cells, which are important for protective adaptive immunity.
Inappropriate activation of these immune cells may cause inammatory disease, and
NFkB inhibitors are promising antiinammatory drug candidates. Here, we inves-
tigated whether inhibiting the NFkBinducing kinase IKK2 can attenuate crescentic
GN, a severe DCand Th celldependent kidney inammatory disease. Prophylactic
pharmacologic IKK2 inhibition reduced DC and Th cell activation and ameliorated
nephrotoxic seruminduced GN in mice. However, therapeutic IKK2 inhibition dur-
ing ongoing disease aggravated the nephritogenic immune response and disease
symptoms. This effect resulted from the renal loss of regulatory T cells, which have
been shown to protect against crescentic GN and which require IKK2. In conclusion,
although IKK2 inhibition can suppress the induction of nephritogenic immune re-
sponses in vivo, it may aggravate such responses in clinically relevant situations,
because it also impairs regulatory T cells and thereby, unleashes preexisting neph-
ritogenic responses. Our ndings argue against using IKK2 inhibitors in chronic GN
and perhaps, other immunemediated diseases.
J Am Soc Nephrol 27: cccccc, 2015. doi: 10.1681/ASN.2015060699
Inhibitor of NFkB kinase subunit 2
(IKK2; also known as IKKb)triggers
the classic NFkB activation pathway,
which is critical for the activation of den-
dritic cells (DCs) and CD4
+
Thelper
(Th) cells during adaptive immune re-
sponses.
1,2
NFkBdependent DC ac-
tivation (for example, in response to
microbial molecular patterns) results in
the upregulation of costimulatory mol-
ecules, such as CD80, CD86, or CD40,
which promote immunogenic Th cell ac-
tivation.
3
IKK2 deletion in T cells pre-
vented their activation and effector func-
tion.
1,4
Thus, the NFkBpathwayis
widely considered to promote inamma-
tion, and various NFkB inhibitors are
currently being tested for the treatment
of immune-mediated and inammatory
diseases.
5
Some IKK2 inhibitors have
shown anti-inammatory effects in pre-
clinical studies on arthritis and pulmo-
nary disease.
6,7
By contrast, the targeted genetic de-
letion of IKK2 in nonimmune cells, such
as keratinocytes or hepatocytes, caused
inammatory disease of skin and liver,
respectively.
8,9
Targeted IKK2 deletion
in DCs suppressed not only their acti-
vation but also, their migration into
draining lymph nodes and their ability
to induce differentiation of regulatory
T(T
reg
) cells.
10
Such T
reg
cells are im-
portant to maintain immunologic self-
tolerance by inhibiting autoreactive T and
B cells and require the forkhead/winged
helix box P3 transcription factor.
1113
They also require classic NFkB pathway
components, including IKK2, for their
development in the thymus.
4,1416
Furthermore, recent studies described
distinct NFkB components with anti-
inammatory properties.
17,18
Thus, there
are both pro- and anti-inammatory
functions of the NFkBpathway,buttheir
interplay and regulation in the in vivo sit-
uation are unclear.
4,19
Received June 25, 2015. Accepted October 8,
2015.
J.G. and E.P. contributed equally to this work.
Present address: Dr. Chen Yao, Organ Transplant
Institute, Chinese PLA 309th Hospital, Beijing, China.
Published online ahead of print. Publication date
available at www.jasn.org.
Correspondence: Prof. Christian Kurts, Institute of
Experimental Immunology, Rheinische Friedrich
Wilhelms University, Sigmund Freud Street 25,
53105 Bonn, Germany, or Prof. Friedrich Thaiss,
Third Medical Department of Clinical Medicine,
University Hospital Hamburg Eppendorf, Martini
Street 52, Hamburg 20246, Germany. Email:
ckurts@web.de or thaiss@uke.de
Copyright © 2015 by the American Society of
Nephrology
J Am Soc Nephrol 27: cccccc, 2015 ISSN : 1046-6673/2707-ccc 1
NFkB activation has also been ob-
served in patients
20
and experimental
models of GN.
21
Crescentic GN (cGN)
is a severe inammatory kidney disease,
which is mediated by Th cells specicfor
glomerular antigens and may rapidly
progress to terminal kidney failure.
22,23
It can be mimicked by the passive neph-
rotoxic nephritis (pNTN) model, which
is induced by injecting a nephrotoxic
sheep antiserum specicformurineglo-
merular components into mice. In the
immune activation phase of this model,
which lasts until days 45afterserum
injection, DCs in lymphatic organs
capture sheep Ig and activate specic
Th cells. In the effector phase starting at
days 34, these Th cells enter the kidney,
where the antiserum is deposited be-
cause of its specicity, and produce
effector cytokines, like IFNg,thatacti-
vate macrophages.
22,23
During their ef-
fector phase, Th cells can be regulated
by kidney-resident DCs, whose activa-
tion state determines whether they stim-
ulate or inhibit the Th cells and thus,
whether nephritis progresses or heals.
24
Here, we hypothesized that inhibiting
DC and Th cell activation with an NFkB
inhibitor should attenuate GN. We tested
this hypothesis by treating mice every
other day with the IKK2 inhibitor kinase
inhibitorof NFkB-1 (KINK-1)
7,25
starting
1 day before administration of the neph-
rotoxic serum (experimental plan is in
Figure 1A). This drug reduced NFkBnu-
clear translocation in vivo (Supplemental
Figure 1) and attenuated pNTN, which
was evidenced by fewer crescents in his-
tologic sections (Figure 1B), lower tubu-
lointerstitial injury (Figure 1C), higher
creatinine clearance (Figure 1D), lower
BUN (Figure 1E), and lower proteinuria
(Figure 1F). Renal DCs (ow cytometric
gating strategy is in Supplemental Figure
2) showed a less activated phenotype
(Figure 1G), and intrarenal activated Th
cells producing IFNgwere less frequent
(Figure 1H). Thus, IKK2 inhibition start-
ing before pNTN induction attenuated
the nephritogenic Th cell response and
disease symptoms.
We noted that antisheep Ig titers,
which can be considered a parameter
for the nephritogenic Th cell response,
were systemically reduced after IKK2 in-
hibition (Figure 1I). Furthermore, DCs
in the spleen appeared less mature (Fig-
ure 1J). This indicated that the induction
of the pNTN model had been compro-
mised; in other words, prophylactic
IKK2 inhibition before pNTN induction
(Figure 1A) had suppressed the induc-
tion of this disease model rather than
preventing its progression. Because pro-
phylactic IKK2 inhibition does not
mimic the clinically relevant situation
of a patient presenting with ongoing dis-
ease, we modied our protocol and ap-
plied the IKK2 inhibitor on days 4, 6, and
8 after disease induction (experimental
plan is in Figure 2A). However, under
these conditions, pNTN was no longer
attenuated (Figure 2, BF), and neither
renal inammation (Figure 2, G and H)
nor systemic antirenal immune response
(Figure 2, I and J) were decreased. This
might indicate that IKK2 inhibition can-
not attenuate ongoing pNTN. Alterna-
tively, the duration of IKK2 inhibition
(on days 4, 6, and 8 compared with six
times treatment in Figure 1) may have
been too short to affect disease.
To distinguish between these possibil-
ities, we decided to use a protocol that
allows prolonged IKK2 inhibition selec-
tively in the Th cell effector phase. Because
these phases overlap between days 3 and 5
in pNTN,
23
we switched to the accelerated
nephrotoxic nephritis (aNTN) model,
wheremicearerst immunized with
sheep Ig to allow for the activation of spe-
cic Th cells without a nephritogenic ef-
fector phase; 5 days later, we injected a
lower dose of nephrotoxic sheep serum
to target these Th cells to the kidney and
applied KINK-1 selectively in the effector
phase (experimental plan is in Figure 3A).
Surprisingly, in this setting, aNTN was
markedly and consistently aggravated,
which was evident by more severe histo-
logic kidney damage (Figure 3, BD, com-
pare the rst two experimental groups),
lower creatinine clearance, more elevated
BUN, and higher proteinuria (Figure 3,
EG). There were more F4/80
+
immune
cells (Figure 3H), intrarenal DCs were
more activated (Figure 3I), and more ac-
tivated IFNg
+
Th cells were detected (Fig-
ure 3J). Antisheep Ig titers as a parameter
for the nephritogenic Th1 cell response
were increased as well (Figure 3K).
Importantly, we noted that intrarenal
T
reg
cells (gating strategy is in Supplemen-
tal Figure 2) were less frequent in both
pNTN and aNTN when the KINK-1 was
given six times (Figure 3, L and N) but
that only a slight and nonsignicant re-
duction was seen in the therapeutic
pNTN setting with only three applications
(Figures 2 and 3M). Because T
reg
cells are
well documented to suppress pNTN and
aNTN
2629
and because the thymic gener-
ation of T
reg
cells requires IKK2,
4,1416
we
hypothesized that KINK-1 might have tar-
geted these cells.
We tested this hypothesis by treating
DEREG mice, in which T
reg
cells can be
conditionally depleted, with KINK-1 (ex-
perimental setting is in Figure 3A, all four
groups). Both T
reg
depletion and IKK2 in-
hibition aggravated aNTN, but no additive
aggravation was seen when these proce-
dures were combined (Figure 3, BG, all
four groups). Likewise, the parameters for
intrarenal inammation (Figure 3, HJ)
and antisheep Ig titers (Figure 3K) were
increased by T
reg
depletion and IKK2
inhibition, but no synergy between these
maneuvers was evident. Thus, therapeutic
IKK2 inhibition aggravated aNTN but was
unable to further increase damage when
T
reg
cells were absent, indicating that
KINK-1 aggravated aNTN by reducing
T
reg
numbers.
Finally, we examined the mechanisms
by which T
reg
cells can suppress immune
responses. After KINK-1 application and
after T
reg
depletion, less TGF-band less
IL-10 was detectable in kidney digests, but
no additive effect of these measures was
noted (Figure 4, A and C, Supplemental
Figure 3). As a control, we measured the
NFkBdependent inammatory cytokine
TNFaand found that it was increased af-
ter both maneuvers but again, not syner-
gistically (Figure 4E, Supplemental Figure
3). Intracellular levels of neither TGF-bor
IL-10 in T
reg
cells (Figure 4, B and D) nor
TNFain DCs or macrophages (Figure 4, F
andG)weresignicantly changed, indi-
cating that KINK-1 acted by reducing ei-
ther the numbers of cytokineproducing
immune cells or hypothetic cytokine pro-
duction by nonimmune cells.
2Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
In summary, our study shows diamet-
rical consequences of prophylactic and
therapeutic in vivo inhibition of IKK2:
prophylactic inhibition prevented DC
and Th cell activation, consistent with the
widely accepted view that the NFkBpath-
way is proinammatory.
2,3,19
Prophylactic
inhibition over at least 1 week suppressed
T
reg
cellsaswell,butthiswasinconsequen-
tial; there was no nephritogenic immune
response that T
reg
cells would have to regu-
late, because the Th effector cells had not
been properly activated. However, when we
applied the IKK2 inhibitor in a realistic dis-
ease situation (i.e., after Th cell activation),
which mimics the situation of patients pre-
senting with ongoing crescentic GN, the
suppression of T
reg
cells was able to unleash
the preexisting nephritogenic Th cell re-
sponse, and disease was aggravated.
We conclude that IKK2 inhibition is
anti-inammatory only when initiated
before disease onset, which is impractical
for clinical use. Therapeutic IKK2 inhibi-
tion has potential proinammatory con-
sequences, because it compromises the
regulation of pathogenic immune re-
sponses through T
reg
cells, and this func-
tion prevails over the anti-inammatory
effect resulting from inhibiting DC
Figure 1. Prophylactic IKK2 inhibition blocks the induction of pNTN. Prophylactic IKK2 inhibition in pNTN. (A) Experimental plan, (B)
representative periodic acidSchiffstained glomeruli and percentage of crescentic glomeruli, (C) tubulointerstitial injury, (D) creatinine
clearance, (E) BUN, (F) albumin-to-creatinine ratio, (G) expression of the DC activation markers CD40 and CD80, (H) numbers of activated
and percentages of IFNgproducing Th cells, (I) IgG titers of antisheep Ig antibodies, and (J) DC activation markers on splenic cells in
nephritic mice prophylactically treated with KINK-1 or vehicle. Data are representative of three independent experiments (n=5; Kruskal
Wallis test with post hoc analysis by MannWhitney test). cGN, crescentic glomerulonephritis; MFI, mean uorescence intensity; NTS,
nephrotoxic serum nephritis. *P,0.05; **P,0.01; ***P,0.001.
J Am Soc Nephrol 27: cccccc, 2015 IKK2 Inhibition in GN 3
www.jasn.org BRIEF COMMUNICATION
maturation. These ndings highlight the
need for in vivo studies in which a com-
plex pathway, such as NFkB, can exert
antagonistic functions in different cell
types or at different time points. Our
ndings imply that great care is neces-
sary in clinical studies aiming to treat
chronic kidney inammation by NFkB
inhibition, at least when IKK2 inhibi-
tors are used, because these might ag-
gravate rather than ameliorate disease.
Our ndings may apply to immune-
mediated diseases affecting other or-
gans as well.
CONCISE METHODS
Mice and Reagents
Mice were bred at the animal facilities of the
University Hospital Bonn and University
Hospital Hamburg Eppendorf under specic
pathogenfree conditions. Animal experiments
Figure 2. Shortterm therapeutic IKK2 inhibition fails to attenuate pNTN. (A) Experimental plan, (B) representative periodic acidSchiff
stained glomeruli and percentage of crescentic glomeruli, (C) tubulointerstitial injury, (D) creatinine clearance, (E) BUN, (F) albumin-
to-creatinine ratio, (G) expression of the DC activation markers CD40 and CD80, (H) numbers of activated and percentages of
IFNgproducing Th cells, (I) IgG titers of antisheep Ig antibodies, and (J) DC activation markers on splenic cells in nephritic mice treated
therapeutically (starting day +4 after the induction of nephrotoxic nephritis) with KINK-1 or vehicle. Data are representative of three in-
dependent experiments (n=4; KruskalWallis test with post hoc analys is by MannWhitney test). cGN, crescent ic glomerulonephritis; MFI,
mean uorescence intensity; NTS, nephrotoxic serum nephritis.
4Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
Figure 3. IKK2 inhibition aggravates aNTN by reducing intrarenal T
reg
cells. (AK) Therapeutic IKK2 inhibition was performed in the
effector phase of accelerated cGN. (A) When indicated, T
reg
cells were depleted using DT in DEREG mice as shown in the experimental
plan. (B) Representative periodic acidSchiffstained glomeruli, (C) percentage of crescentic glomeruli, (D) tubulointerstitial injury, (E)
creatinine cleara nce, (F) BUN, (G) albumin-to-cr eatinine ratio, (H) number of F4/8 0
+
immune cells counte d on sections per high-power eld
(hpf), (I) expression of the DC activation markers CD40 and CD80, (J) numbers of activated and percentages of IFNgproducing Th cells,
and (K) serum antisheep IgG titers. (LN) Number of intrarenal T
reg
cells in the experiments shown in Figures 1 and 2 and AK, respectively.
Data are representative of four independent experiments (n=5; post hoc analysis by ANOVA with Bonferroni post-test). cGN, crescentic
glomerulonephritis; DT, diphtheria toxin; MFI, mean uorescence intensity; NTS, nephrotoxic serum. *P,0.05; **P,0.01; ***P,0.001.
J Am Soc Nephrol 27: cccccc, 2015 IKK2 Inhibition in GN 5
www.jasn.org BRIEF COMMUNICATION
were performed according to national and insti-
tutional animal care and ethical guidelines and
had been approved by government committees
(Behörde für Gesundheit und Verbraucherschutz
BGV der Freien und Hansestadt Hamburg
and Landesamt für Natur and Umwelt und
Verbraucherschutz Nordrhein-Westfalen).
T
reg
cells were depleted in nephritic DEREG
micebyinjecting15ng/gbodywtdiphtheria
toxin.
27,28
KINK-1 is an ATPcompetitive selec-
tive IKK2 kinase inhibitor.
7
KINK-1 was dis-
solved in 10% cremophor (Sigma-Aldrich,
St. Louis, MO) diluted with PBS. A KINK-1
dose of 5 mg/kg body wt subcutaneously every
second day inhibited IKK2 in mice.
pNTN and aNTN Models
pNTN was induced in 8- to 10-week-old age
and sexmatched male mice (2025 g body
wt; Charles River Laboratories, Wilmington,
MA) on the C57BL/6J background by intra-
peritoneal injection of 2.5 mg/kg body wt
sheep antimouse glomerular basement
membrane antiserum (nephrotoxic nephritis
serum) as described.
24
Controls received
an equal amount of nonspecic sheep IgG.
In aNTN, mice were injected on day 25
with 2.5 mg/kg body wt sheep IgG in incom-
plete Freunds adjuvant subcutaneously and
day 0 with 2 mg/kg body wt nephrotoxic ne-
phritis serum intraperitoneally. This dose
caused intermediate disease severity, and doses
$2.5 mg/kg body wt caused complete renal fail-
ure after 2 weeks. Urine was collected in metabolic
cages for 24 hours, and serum was derived from
whole blood after cardiac puncture. Urinary cre-
atinine, serum creatinine, and BUN were mea-
sured using standard methods in the central
laboratory of Bonn University Hospital.
24
Albuminuria (Mice Albumin Kit; Bethyl) and se-
rum IgG (Dianova) were determined by ELISA.
Histology
Renal tissue injury was assessed in 2-mm
paraformaldehyde (4%)xed parafn tissue
sections stained by periodic acidSchiff reac-
tion. A semiquantitative score for acute glo-
merular injury was assessed in 30 glomeruli
per mouse by a double-blinded observer as
described before.
28
Kidney damage was his-
tologically scored by an observer blinded to
the identity of samples. For determining
the proportion of crescentic glomeruli,
$80 glomeruli per section were examined.
F4/80
+
cells were stained using rabbit anti-
body to F4/80 (MCA497B; Serotec) diluted
1:50. F4/80
+
cell inltration was then quanti-
ed using ImageJ software (National Institutes
of Health). Light microscopic evaluation was
performed under an Axioskop (Carl Zeiss
GmbH, Jena, Germany) and photographed
with an Axiocam HRc (Carl Zeiss GmbH)
using the Axiostar software (Carl Zeiss
GmbH). F4/80
+
cells in 30 tubulointerstitial
highpower elds per kidney were counted
by light microscopy.
Flow Cytometry
Complete kidneys and spleens were digested
with collagenase (Roche Diagnostics, Indian-
apolis, IN) and DNAse-I as previously de-
scribed.
30
Single-cell suspensions were
stained with uorochrome-conjugated anti-
bodies in PBS containing 10% FCS. The fol-
lowing antibodies from BD Pharmingen or
eBioscience (San Diego, CA) were used:
CD4 (GK1.5), anti-CD45 (30F11), CD8
(536.7), B220 (RA36B2), CD25 (PC61.5),
forkhead/wingedhelix box P3 (FJK-16S),
CD11c (HL3), MHC-II (M5/114.15.2),
F4/80 (BM8), Gr1 (RB68C5), CD40 (3/23),
CD69 (1H.2F3), CD80 (1610A1), and anti-
IFNg(XMG1.2). Dead cells were excluded
by staining with the LIVE/DEAD Fixable
Violet Dead Cell Stain Kit. Viable CD11c
+
MHC II
+
cells were considered DCs. Intra-
cellular staining was performed as recently
described.
13
Cells were analyzed with a BD
Biosciences (San Jose, CA) LSRII using Diva
and FlowJo software. Kidney singlecell sus-
pensions were overnight restimulated with
25 mg/ml sheep Ig, and then, concentrations
of TGF-b, IL-10, and TNFawere measured
Figure 4. Intrarenal cytokine milieu after IKK2 inhibition in aNTN. (A and B) TGF-b, (C and D) IL-10, and (EG) TNFalevels in (A, C, and E)
whole-kidney digests measured by ELISA or Luminex and (B and D) T
reg
cells, (F) DCs, and (G) macrop hages measured by intracellular ow
cytometry. Data are representative of two independent experiments (n=4; post hoc analysis by ANOVA with Bonferroni post-test). cGN,
crescentic glomerulonephritis; DT, diphtheria toxin; MFI, mean uorescence intensity. *P,0.05.
6Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
by ELISA or Luminex according to the man-
ufacturers instructions (eBioscience).
Nuclear Fractionation and Gel Shift
Experiment
Kidneys were perfused with 50 ml sterile PBS
per animal before harvesting. Nuclear mini-
ature extracts were prepared, and gel shift
assays were carried out using an NFkB oligo-
nucleotide probe (Promega, Heidelberg,
Germany) end labeled with
32
P-g-ATP
(3000 Ci/mmol; GE Healthcare, Waukesha,
WI). Afterward, 30 mg nuclear protein was
incubated for 30 minutes at room tempera-
ture with 100,000 cpm probe in 20 mM
HEPES (pH 7.9), 0.3 mM EDTA, 0.2 mM
EGTA, 80 mM NaCl, and 2 mg poly(dI-dC)
poly(Di-dC) (Amersham Pharmacia Biotech)
in a total volume of 20 ml. Where indicated,
competition experiments were performed by
adding unlabeled consensus oligonucleotides
in a 100-fold molar excess to the binding re-
action. The DNA-protein complexes were
separated by electrophoresis and autoradio-
graphed at 280°C for 1 week. Exposed lms
were quantied using a phosphoimager Bio-
Rad GS-363 (multianalyst software; Hercules,
CA) and corrected to the density of the probe.
Statistical Analyses
Results are expressed as me ans6SEM. Differ-
ences between experimental groups were
compared by either the KruskalWallis test
with post hoc analysis usingthe MannWhitney
test or one-way ANOVA with post hoc Bonferroni
test of selected groups (GraphPad Prism
Software; GraphPad Software, La Jolla, CA).
Paired ttest was used to compare mean values
within one experimental series. No random-
ization or exclusion of data points was used.
Statistical signicance was dened as P,0.05.
Experiments yielding insufcient data for sta-
tistical analysis because of the experimental
setup were repeated at least three times. Tests
were reported only where data met assumptions
of tests. On the basis of preliminary experimental
data, a power analysis of 0.8 with P,0.05
indicates a minimum number of three samples/
purications per group, but in some cases, four
samples/purications per group were used.
ACKNOWLEDGMENTS
We thank Chrystel Flores and Anna Kaffke
for excellent technical assistance and Tim
Sparwasser for DEREG mice. C.K. is a member
of the Excellence Cluster ImmunoSensation.
We acknowledge technicalsupport from the
Central Animal Facilities and the Flow Cy-
tometry Core Facilitiesof the Medical Faculties
both in Bonn and in Hamburg. This work was
funded by Deutsche Forschungsgemeinschaft
Grants KFO228 and SFBTR57, Gottfried
Wilhelm Leibniz Price (to C.K.), and the
European Union Consortia INTRICATE and
RELENT.
DISCLOSURES
K.Z. is a full-time employee and stockholder of
Bayer AG (Berlin, Germany). The other authors
declare no competing nancial interest.
REFERENCES
1. Oh H, Ghosh S: NF-kB: Roles and regulation
in different CD4(+) T-cell subsets. Immunol
Rev 252: 4151, 2013
2. Vallabhapurapu S, Karin M: Regulation and
function of NF-kappaB transcription factors
in the immune system. Annu Rev Immunol
27: 693733, 2009
3. Steinman RM, Bonifaz L, Fujii S, Liu K,
Bonnyay D, Yamazaki S, Pack M, Hawiger D,
Iyoda T, Inaba K, Nussenzweig MC: The in-
nate functions of dendritic cells in peripheral
lymphoid tissues. Adv Exp Med Biol 560: 83
97, 2005
4. Gerondakis S, Fulford TS, Messina NL,
Grumont RJ: NF-kB control of T cell devel-
opment. Nat Immunol 15: 1525, 2014
5. Gasparini C, Feldmann M: NF-kBasatarget
for modulating inam matory responses. Curr
Pharm Des 18: 57355745, 2012
6. McIntyre KW, Shuster DJ, Gillooly KM,
Dambach DM, Pattoli MA, Lu P, Zhou XD,
Qiu Y, Zusi FC, Burke JR: A highly selective
inhibitor of I kappa B kinase, BMS-345541,
blocks both joint inammation and destruc-
tion in collagen-induced arthritis in mice.
Arthritis Rheum 48: 26522659, 2003
7. Ziegelbauer K, Gantner F, Lukacs NW, Berlin
A, Fuchikami K, Niki T, Sakai K, Inbe H,
Takeshita K, Ishimo ri M, Komura H, Murata T,
Lowinger T, Bacon KB: A selective novel
low-molecular-weight inhibitor of IkappaB
kinase-beta (IKK-beta) prevents pulmo-
nary inammation and shows broad anti-
inammatory activity. Br J Pharmacol 145:
178192, 2005
8. Pasparakis M, Haase I, Nestle FO: Mecha-
nisms regulating skin immunity and inam-
mation. Nat Rev Immunol 14: 289301,
2014
9. Malato Y, Sander LE, Liedtke C, Al-Masaoudi
M, Tacke F, Trautwein C, Beraza N: Hepatocyte-
specic inhibitor-of-kappaB-kinase deletion
triggers the innate immune response and
promotes earlier cell proliferation during liver
regeneration. Hepatology 47: 20362050,
2008
10. Baratin M, Foray C, Demaria O, Habbeddine
M, Pollet E, Maurizio J, Verthuy C, Davanture
S, Azukizawa H, Flore s-Langarica A, Dalod M,
Lawrence T: Homeostatic NF-kB signaling in
steady-state migratory dendritic cells regu-
lates immune homeostasis and tolerance.
Immunity 42: 627639, 2015
11. Gavin MA, Rasmussen JP, Fontenot JD,
Vasta V, Manganiello VC, BeavoJA, Rudensky
AY: Foxp3-dependent programme of regu-
latory T-cell differentiation. Nature 445:
771775, 2007
12. Sakaguchi S, Yamaguchi T, Nomura T, Ono
M: Regulatory T cells and immune tolerance.
Cell 133: 775787, 2008
13. Gotot J, Gottschalk C, Leopold S, Knolle PA,
Yagita H, Kurts C, Ludwig-Portugall I: Regu-
latory T cells use programmed death 1 li-
gands to directly suppress autoreactive
B cells in vivo. Proc Natl Acad Sci U S A 109:
1046810473, 2012
14. Schmidt-Supprian M, Tian J, Grant EP,
Pasparakis M, Maehr R, Ovaa H, Ploegh HL,
Coyle AJ, Rajewsky K: Differential de-
pendence of CD4+CD25+ regulatory and
natural killer-like T cells on signals leading to
NF-kappaB activation.Proc Natl Acad Sci U S A
101: 45664571, 2004
15. Long M, Park SG, Strickland I, Hayden MS,
Ghosh S: Nuclear factor-kappaB modulates
regulatory T cell development by directly
regulating expression of Foxp3 transcription
factor. Immunity 31: 921931, 2009
16. Gückel E, Frey S, Zaiss MM, Schett G, Ghosh
S, Voll RE: Cell-intrinsic NF-kB activation
is criti cal for the d evelopment of natural
regulatory T cells in mice.PLoS One 6: e20003,
2011
17. Dissanayake D, Hall H, Berg-Brown N, Elford
AR, Hamilton SR, Murakami K, Deluca LS,
Gommerman JL, Ohashi PS: Nuclear factor-
kB1 controls the functional maturation of
dendritic cells and prevents the activation
of autoreactive T cells. Nat Med 17: 1663
1667, 2011
18. Brüstle A, Brenner D, Knobbe CB, Lang PA,
Virtanen C, Hersheneld BM, Reardon C,
Lacher SM, Ruland J, Ohashi PS, Mak TW:
The NF-kB regulator MALT1 determines the
encephalitogenic potential of Th17 cells. J
Clin Invest 122: 46984709, 2012
19. Hayden MS, Ghosh S: NF-kB, the rst quarter-
century: Remarkable progress and outstand-
ing questions. Genes Dev 26: 203234,
2012
20. Silva GE, Costa RS, Ravinal RC, Ramalho LZ,
Dos Reis MA, Coimbra TM, Dantas M: NF-kB
expression in IgA nephropathy outcome. Dis
Markers 31: 915, 2011
21. Brähler S, Ising C, Hagmann H, Rasmus M,
Hoehne M, Kurschat C, Kisner T, Goebel H,
Shankland S, Addicks K, Thaiss F, Schermer
J Am Soc Nephrol 27: cccccc, 2015 IKK2 Inhibition in GN 7
www.jasn.org BRIEF COMMUNICATION
B, Pasparakis M, Benzing T, Brinkkoetter PT:
Intrinsic proinammatory signaling in podo-
cytes contributes to podocyte damage and
prolonged proteinuria. Am J Physiol Renal
Physiol 303: F1473F1485, 2012
22. Tipping PG, Holdsworth SR: T cells in cres-
centic glomerulo nephritis. J Am Soc Nephrol
17: 12531263, 2006
23. Kurts C, Panzer U, Anders HJ, Rees AJ: The
immune system and kidney disease: Basic
concepts and clinical implications. Nat Rev
Immunol 13: 738753, 2013
24. Hochheiser K, Heuser C, Krause TA, Teteris
S, Ilias A, Weisheit C, Ho ssF , Tittel AP, Knolle
PA, Panzer U, Engel DR, Tharaux PL, Kurts C:
Exclusive CX3CR1 dependence of kidney
DCs impacts glomerulonephritis progres-
sion. JClinInvest123: 42424254, 2013
25. S chön M, Wienrich BG, Kneitz S, Sennefeld er
H, Amschler K, Vöhrin ger V, Weber O, Stiewe
T,ZiegelbauerK,SchönMP:KINK-1,anovel
small-molecule inhibitor of IKKbeta, and the
susceptibility of melanoma cells to antitu-
moral treatment. J Natl Cancer Inst 100:
862875, 2008
26. Wolf D, Hochegger K, Wolf AM, Rumpold
HF, Gastl G, Tilg H, Mayer G, Gunsilius E,
Rosenkranz AR: CD4+CD25+ regulatory
T cells inhibit experimental anti-glomerular
basement membrane glomerulonephritis in
mice. J Am Soc Nephrol 16: 13601370, 2005
27. Ooi JD, Snelgrove SL, Engel DR, Hochheiser K,
Ludwig-Portugall I, Nozaki Y, OSullivan KM,
Hickey MJ, Holdsworth SR, Kurts C, Kitching AR:
Endogenous foxp3(+) T-regulatory cells sup-
press anti-glomerular basement membrane
nephritis. Kidney Int 79: 977986, 2011
28. Paust HJ, Ostmann A, Erhardt A, Turner JE,
Velden J,Mittrücker HW, SparwasserT, Panzer
U, Tiegs G: Regulatory T cells control the Th1
immune response in murine crescentic glo-
merulonephritis. Kidney Int 80: 154164, 2011
29. Ostmann A, Paust HJ, Panzer U, Wegscheid
C, Kapffer S, Huber S, Flavell RA, Erhardt A,
Tiegs G: Regulatory T cell-derived IL-10
ameliorates crescentic GN. JAmSocNeph-
rol 24: 930942, 2013
30. Krüger T, Benke D, Eitner F, Lang A, Wirtz M,
Hamilton-Williams EE, Engel D, Giese B,
Müller-Newen G, Floege J, Kurts C: Identi-
cation and functional characterization of
dendritic cells in the healthy murine kidney
and in experimental glomerulonephritis.
J Am Soc Nephrol 15: 613621, 2004
This article contains supplemental material online at
http://jasn.asnjournals.org/lookup/suppl/doi:10.1681/
ASN.2015060699/-/DCSupplemental.
8Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
... A study published from our laboratory by Gotot et al. (20) showed that systemic IKK-2 inhibition after the induction of the disease increased renal injury in the NTN model and that activated cytotoxic T lymphocytes were resistant to IKK-2 inhibition because of other pathways-in particular, nuclear factor of activated T-cell signaling (21). NF-kB activation was demonstrated by our group after ischemia-reperfusion kidney injury, and more Th17 cells were found in ischemic CD4xNEMO D mice but not in CD4xIKK2 D mice (22). ...
... We and others have demonstrated NF-kB activation in glomerulonephritis, and in 2016 we demonstrated that IKK-2 inhibition before NTN induction attenuated the nephritogenic T h cell response (20). Therefore, genetically modified mice were used to examine the effects of IKK-2 or NEMO deletion, specifically in T lymphocytes, in the experimental NTN model. ...
... We have shown previously that total kidney NF-kB was activated after the induction of NTN (20), which was also confirmed in CD4xIKK2 D and CD4xNEMO D mice and may explain why no significant functional and morphologic changes were observed at 10 d after NTN induction. Our results do not exclude, however, that functional or morphologic changes may occur at later time points and influence the long-term outcome in these mice. ...
Article
Full-text available
Experimental nephrotoxic serum nephritis (NTN) is a model for T‐cell–mediated human rapid progressive glomerulonephritis. T‐cell receptor stimulation involves intracellular signaling events that ultimately lead to the activation of transcription factors, such as NF‐κB. We explored the involvement of the NF‐κB components IKK‐2 and NEMO in NTN, by using cell‐specific knockouts of IKK‐2 and NEMO in CD4⁺ T lymphocytes. Our results demonstrate that although the course of disease was not grossly altered in CD4xIKK2Δ and CD4xNEMOΔ animals, renal regulatory T cells were significantly reduced and T helper (Th)1 and Th17 cells significantly increased in both knockout mouse groups. The expression of the renal cytokines and chemokines IL‐1β, CCL‐2, and CCL‐20 was also significantly altered in both knockout mice. Lymphocyte transcriptome analysis confirmed the increased expression of Th17‐related cytokines in spleen CD4⁺ T cells. Moreover, our array data demonstrate an interrupted canonical NF‐κB pathway and an increased expression of noncanonical NF‐κB pathway–related genes in nephritic CD4xNEMOΔ mice, highlighting different downstream effects of deletion of IKK‐2 or NEMO in T lymphocytes. We propose that better understanding of the role of IKK‐2 and NEMO in nephritis is essential for the clinical application of kinase inhibitors in patients with glomerulonephritis.—Guo, L., Huang, J., Chen, M., Piotrowski, E., Song, N., Zahner, G., Paust, H.‐J., Alawi, M., Geffers, R., Thaiss, F. T‐lymphocyte–specific knockout of IKK‐2 or NEMO induces Th17 cells in an experimental nephrotoxic nephritis mouse model. FASEB J. 33, 2359–2371 (2019). www.fasebj.org
... We recently observed that prolonged treatment with the IKKb inhibitor KINK-1 (kinase inhibitor of NF-kB-1) surprisingly aggravated a T helper (Th) cell-mediated kidney disease model (Gotot et al., 2016). Given that NF-kB activation promotes FoxP3 expression (Ruan et al., 2009;Schuster et al., 2012;Zheng et al., 2010) and that CD25 signaling promotes Treg survival (Furtado et al., 2002), we hypothesized that mature Tregs may require IKKb for expansion, maintenance, and/or recruitment. ...
... We recently noted lower Treg numbers after prolonged IKKb inhibition in a kidney disease model (Gotot et al., 2016). To investigate whether Tregs require cell-intrinsic IKKb, we crossed IKKb fl/fl mice (Park et al., 2002) with FoxP3 Cre mice (Rubtsov et al., 2008) to generate mice whose FoxP3-expressing cells lacked IKKb (termed FoxP3 DIKKb mice). ...
... Flow cytometric analysis after 15 days showed almost 50% less splenic FoxP3 + Tregs, whereas Th cells, CTLs ( Figure 2F), and total numbers of circulating leukocytes (data not shown) remained normal, indicating that systemic IKKb inhibition acted preferentially on Tregs. DCs and macrophages showed elevated signs of activation ( Figure S2B), presumably because of the loss of Tregs, consistent with our previous study on the effects of KINK-1 in myeloid cells (Gotot et al., 2016). Scurfy symptoms were not observed under KINK-1 treatment, consistent with the reduction of Tregs by only 50%. ...
Article
Full-text available
Regulatory T cells (Tregs) prevent autoimmunity but limit antitumor immunity. The canonical NF-κB signaling pathway both activates immunity and promotes thymic Treg development. Here, we report that mature Tregs continue to require NF-κB signaling through IκB-kinase β (IKKβ) after thymic egress. Mice lacking IKKβ in mature Tregs developed scurfy-like immunopathology due to death of peripheral FoxP3⁺ Tregs. Also, pharmacological IKKβ inhibition reduced Treg numbers in the circulation by ∼50% and downregulated FoxP3 and CD25 expression and STAT5 phosphorylation. In contrast, activated cytotoxic T lymphocytes (CTLs) were resistant to IKKβ inhibition because other pathways, in particular nuclear factor of activated T cells (NFATc1) signaling, sustained their survival and expansion. In a melanoma mouse model, IKKβ inhibition after CTL cross-priming improved the antitumor response and delayed tumor growth. In conclusion, prolonged IKKβ inhibition decimates circulating Tregs and improves CTL responses when commenced after tumor vaccination, indicating that IKKβ represents a druggable checkpoint.
... However, our current experiments show that IKK1-inhibition in lymphocytes might be a candidate to be evaluated further as a treatment option in kidney diseases. We and others have shown previously that the activation of T cells with suppressive function, Tregs, is critical depend on the activation of IKK2 so that the activation of IKK1 and IKK2 play critical roles in different T-cell subset activation [74][75][76][77] and therefore might be therapeutic targets to inhibit or activate different T-cell subpopulations at different time points as kidney diseases progress. ...
Article
Full-text available
Ischemia–reperfusion injury (IRI) is one of the major causes of acute kidney injury (AKI), and experimental work has revealed detailed insight into the inflammatory response in the kidney. T cells and NFκB pathway play an important role in IRI. Therefore, we examined the regulatory role and mechanisms of IkappaB kinase 1 (IKK1) in CD4⁺T lymphocytes in an experimental model of IRI. IRI was induced in CD4cre and CD4IKK1Δ mice. Compared to control mice, conditional deficiency of IKK1 in CD4⁺T lymphocyte significantly decreased serum creatinine, blood urea nitrogen (BUN) level, and renal tubular injury score. Mechanistically, lack in IKK1 in CD4⁺T lymphocytes reduced the ability of CD4 lymphocytes to differentiate into Th1/Th17 cells. Similar to IKK1 gene ablation, pharmacological inhibition of IKK also protected mice from IRI. Together, lymphocyte IKK1 plays a pivotal role in IRI by promoting T cells differentiation into Th1/Th17 and targeting lymphocyte IKK1 may be a novel therapeutic strategy for IRI. Supplementary Information The online version contains supplementary material available at 10.1007/s00018-023-04763-2.
... Another example can be found in the process of dendritic cell maturation during experimental cGN, which is generally mediated by the transcription factor nuclear factor-κB (NF-κB). In murine cGN, pharmacological inhibition of NF-κB diminished the maturation of DCs, but the subsequent loss of regulatory T cells exacerbated multiple features of crescentic disease (Gotot et al. 2016). ...
Article
Full-text available
Crescentic glomerulonephritis represents a group of kidney diseases characterized by rapid loss of kidney function and the formation of glomerular crescents. While the role of the immune system has been extensively studied in relation to the development of crescents, recent findings show that parietal epithelial cells play a key role in the pathophysiology of crescent formation, even in the absence of immune modulation. This review highlights our current understanding of parietal epithelial cell biology and the reported physiological and pathological roles that these cells play in glomerular lesion formation, especially in the context of crescentic glomerulonephritis.
... TANK-binding kinase 1 (TBK1) can suppress the production of IgA in B cells by accelerating the degradation of NIK, which plays an important role in the NFκB noncanonical pathway (68,69). Our data showed that in crescentic GN, an inhibitor of IKK2, unexpectedly accelerated the disease, which may have resulted from Treg cell impairment (70). In another experimental model of crescentic GN, deficiency in NFκB1 (p50) increased the severity of acute glomerular injury but NFκB1 did not influence the chronic, fibrotic phase (71). ...
Article
Full-text available
The global burden of chronic kidney disease will increase during the next century. As NFκB, first described more than 30 years ago, plays a major role in immune and non-immune-mediated diseases and in inflammatory and metabolic disorders, this review article summarizes current knowledge on the role of NFκB in in vivo kidney injury and describes the new and so far not completely understood crosstalk between canonical and non-canonical NFκB pathways in T-lymphocyte activation in renal disease.
... Subsequently, antibodies against foreign IgG and T cells accumulate in the kidney and cause crescentic GN within 1-2 wk. NTN is associated with renal Treg cell accumulation, and mice with depleted or functionally impaired Treg cell populations develop aggravated crescentic GN (6,9,17,21,23,26,27,29,36). On the other hand, mice with increased Treg cell populations are protected (38,40), which is also the case when Treg cells are expanded by anti-IL-2/IL-2 treatment (Fig. 7). ...
Article
Anti-glomerular basement membrane (anti-GBM) disease is characterized by antibodies and T cells directed against the Goodpasture antigen, the noncollagenous domain of the α3-chain of type IV collagen [α3(IV)NC1] of the GBM. Consequences are the deposition of autoantibodies along the GBM and the development of crescentic glomerulonephritis (GN) with rapid loss of renal function. Forkhead box protein P3 (Foxp3)+ regulatory T (Treg) cells are crucial for the maintenance of peripheral tolerance to self-antigens and the prevention of immunopathology. Here, we use the mouse model of experimental autoimmune GN to characterize the role of Treg cells in anti-GBM disease. Immunization of DBA/1 mice with α3(IV)NC1 induced the formation of α3(IV)NC1-specific T cells and antibodies and, after 8-10 wk, the development of crescentic GN. Immunization resulted in increased frequencies of peripheral Treg cells and renal accumulation of these cells in the stage of acute GN. Depletion of Treg cells during immunization led to enhanced generation of α3(IV)NC1-specific antibodies and T cells and to aggravated GN. In contrast, depletion or expansion of the Treg cell population in mice with established autoimmunity had only minor consequences for renal inflammation and did not alter the severity of GN. In conclusion, our results indicate that in anti-GBM disease, Treg cells restrict the induction of autoimmunity against α3(IV)NC1. However, Treg cells are inefficient in preventing crescentic GN after autoimmunity has been established.
... 99 However, in a mouse model of crescentic GN, inhibition of NF-kB prior to disease induction resulted in improved disease phenotype, but treatment after onset of disease resulted in marked worsening of disease with higher proteinuria, lower creatinine clearance, and greater severity of histology. 100 Further work investigating the role of this complex pathway is needed before translation to clinical studies. ...
Article
Full-text available
IgA nephropathy (IgAN) is the most common form of primary glomerulonephritis worldwide, exhibiting a variable clinical and pathological course and significantly contributing to the global burden of chronic kidney disease and end-stage renal disease. Current standards of care focus on optimization of antihypertensive and antiproteinuric therapies (typically renin- angiotensin system blockade) to reduce disease progression. Much recent attention has focused on whether additional immunosuppression confers better outcomes than supportive management alone, and indeed, several trials have demonstrated renoprotective effects following the use of oral corticosteroids. However, results have been inconsistent, and perceived benefits must be balanced against risks and adverse effects associated with generalized immunosuppression, as highlighted by the high-profile STOP-IgAN and TESTING clinical trials. Recent translational research in vitro and animal models of IgAN have generated greater insight into potential therapeutic targets for this complex autoimmune disease. Deeper understanding of the roles of the mucosal immune barrier, complement activation and deposition, T-cell dependent and independent mechanisms of B cell activation, and of the deposition and downstream inflammatory signaling pathways of nephritogenic polymeric IgA1 complexes (e.g., signaling of immune receptors via spleen tyrosine kinase) has formed the rationale for the development of novel agents and clinical trials of more targeted therapies. However, translating findings into clinical practice is challenging, with many immunopathological features of IgAN specific to humans. Recent comprehensive reviews outline current understanding of mechanisms of IgAN as well as ongoing and future clinical trials; it is not our aim to replicate this here. Instead, we take a mechanistic approach to current treatment strategies, outlining advantages and limitations of each before exploring ongoing research with potential translation into future targeted therapies for this complex disease.
Article
Dendritic cells (DCs) are chief inducers of adaptive immunity and regulate local inflammatory responses across the body. Together with macrophages, the other main type of mononuclear phagocyte, DCs constitute the most abundant component of the intrarenal immune system. This network of functionally specialized immune cells constantly surveys its microenvironment for signs of injury or infection, which trigger the initiation of an immune response. In the healthy kidney, DCs coordinate effective immune responses, for example, by recruiting neutrophils for bacterial clearance in pyelonephritis. The pro-inflammatory actions of DCs can, however, also contribute to tissue damage in various types of acute kidney injury and chronic glomerulonephritis, as DCs recruit and activate effector T cells, which release toxic mediators and maintain tubulointerstitial immune infiltrates. These actions are counterbalanced by DC subsets that promote the activation and maintenance of regulatory T cells to support resolution of the immune response and allow kidney repair. Several studies have investigated the multiple roles for DCs in kidney homeostasis and disease, but it has become clear that current tools and subset markers are not sufficient to accurately distinguish DCs from macrophages. Multidimensional transcriptomic analysis studies promise to improve mononuclear phagocyte classification and provide a clearer view of DC ontogeny and subsets.
Article
Scope: Multiple sclerosis (MS) is an inflammatory demyelinating autoimmune disorder, with increasing incidence worldwide but unknown etiology. 6-gingerol (6-GIN), a major dietary compound found in ginger rhizome, has immunomodulatory activity. However, its role in autoimmune diseases, as well as the underlying mechanisms, are unclear. In this study, we evaluate if 6-GIN can effectively ameliorated the clinical disease severity of experimental autoimmune encephalomyelitis, an animal model of MS. Methods and results: Clinical scores of Experimental autoimmune encephalomyelitis (EAE) mice were recorded daily, inflammation of periphery and neuroinflammation of EAE mice were determined by flow cytometry analysis, ELISA and histopathological analysis, results showed that 6-GIN significantly inhibited inflammatory cell infiltration from the periphery into the central nervous system, and reduced neuroinflammation and demyelination. Flow cytometry analysis, ELISA and quantitative PCR showed that 6-GIN could suppress lipolysaccharide-induced DC activation and induce the tolerogenic DCs. Immunoblot analysis reveals that the phosphorylation of nuclear factor-κB and mitogen-activated protein kinase, two critical regulators of inflammatory signaling, were significantly inhibited in 6-GIN-treated DCs. Conclusion: The results of this study demonstrate that 6-GIN has significant potential as a novel anti-inflammatory agent for the treatment of autoimmune diseases such as MS via direct modulatory effects on DCs. This article is protected by copyright. All rights reserved.
Article
Full-text available
Some studies have demonstrated the involvement of nuclear factor-kappa B (NF-kB) in the pathogenesis of glomerulonephritis. The aim of our study was twofold: (1) to analyze the prognostic value of NF-kB expression in primary IgA nephropathy (IgAN) and (2) to compare the results of NF-kB expression by immunohistochemistry (IHC) and southwestern histochemistry (SWH). We analyzed 62 patients diagnosed with IgAN from 1987 to 2003. We used monoclonal antibodies to CD68 and mast cell tryptase and polyclonal antibodies to TGF-β1, α-SMA and NF-kB p65. We used SWH for the in situ detection of activated NF-kB. The results showed that NF-kB expression (mainly by SWH) correlated with clinical and histological parameters. An unfavorable clinical course of IgAN was significantly related to tubular NF-kB expression by SWH, but not by IHC. The Kaplan-Meier curves demonstrated that increased NF-kB expression, which was measured by IHC and SWH, decreased renal survival. In conclusion, the increased expression of NF-kB in the tubular area may be a predictive factor for the poor prognosis of patients with IgAN. Compared with IHC, NF-kB expression determined by SWH was correlated with a larger number of parameters of poor disease outcome.
Article
Full-text available
The kidneys are frequently targeted by pathogenic immune responses against renal autoantigens or by local manifestations of systemic autoimmunity. Recent studies in rodent models and humans have uncovered several underlying mechanisms that can be used to explain the previously enigmatic immunopathology of many kidney diseases. These mechanisms include kidney-specific damage-associated molecular patterns that cause sterile inflammation, the crosstalk between renal dendritic cells and T cells, the development of kidney-targeting autoantibodies and molecular mimicry with microbial pathogens. Conversely, kidney failure affects general immunity, causing intestinal barrier dysfunction, systemic inflammation and immunodeficiency that contribute to the morbidity and mortality of patients with kidney disease. In this Review, we summarize the recent findings regarding the interactions between the kidneys and the immune system.
Article
Full-text available
DCs and macrophages both express the chemokine receptor CX3CR1. Here we demonstrate that its ligand, CX3CL1, is highly expressed in the murine kidney and intestine. CX3CR1 deficiency markedly reduced DC numbers in the healthy and inflamed kidney cortex, and to a lesser degree in the kidney medulla and intestine, but not in other organs. CX3CR1 also promoted influx of DC precursors in crescentic glomerulonephritis, a DC-dependent aggressive type of nephritis. Disease severity was strongly attenuated in CX3CR1-deficient mice. Primarily CX3CR1-dependent DCs in the kidney cortex processed antigen for the intrarenal stimulation of T helper cells, a function important for glomerulonephritis progression. In contrast, medullary DCs played a specialized role in inducing innate immunity against bacterial pyelonephritis by recruiting neutrophils through rapid chemokine production. CX3CR1 deficiency had little effect on the immune defense against pyelonephritis, as medullary DCs were less CX3CR1 dependent than cortical DCs and because recruited neutrophils produced chemokines to compensate for the DC paucity. These findings demonstrate that cortical and medullary DCs play specialized roles in their respective kidney compartments. We identify CX3CR1 as a potential therapeutic target in glomerulonephritis that may involve fewer adverse side effects, such as impaired anti-infectious defense or compromised DC functions in other organs.
Article
Full-text available
Regulatory T cells (Tregs) exert their immunosuppressive activity through several immunoregulatory mechanisms, including the production of anti-inflammatory cytokines such as IL-10. Although several studies suggest a role for Tregs in modulating crescentic GN, the underlying mechanisms are not well understood. Here, using IL-10 reporter mice, we detected IL-10-producing Foxp3(+) T cells in the kidney, blood, and secondary lymphoid tissue in a mouse model of crescentic GN. Specific inactivation of Il10 in Foxp3(+) Tregs eliminated the ability of these cells to suppress renal and systemic production of IFNγ and IL-17; these IL-10-deficient Tregs lost their capacity to attenuate renal tissue injury. These data highlight the suppressive functions of Tregs in crescentic GN and suggest the importance of Treg-derived IL-10 in ameliorating disease severity and in modulating both the Th1 and most notably Th17 immune response.
Article
Full-text available
Authors: A.B. and D.B. contributed equally to this work. Effector functions of inflammatory IL-17-producing Th (Th17) cells have been linked to autoimmune diseases such as experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). However, what determines Th17 cell encephalitogenicity is still unresolved. Here, we show that after EAE induction, mice deficient for the NF-κB regulator MALT1 (Malt1-/- mice) exhibit strong lymphocytic infiltration in the CNS, but do not develop any clinical signs of EAE. Loss of Malt1 interfered with expression of the Th17 effector cytokines IL-17 and GM-CSF both in vitro and in vivo. In line with their impaired GM-CSF secretion, Malt1-/- Th cells failed to recruit myeloid cells to the CNS to sustain neuroinflammation, whereas autoreactive WT Th cells successfully induced EAE in Malt1-/- hosts. In contrast, Malt1 deficiency did not affect Th1 cells. Despite their significantly decreased secretion of Th17 effector cytokines, Malt1-/- Th17 cells showed normal expression of lineage-specific transcription factors. Malt1-/- Th cells failed to cleave RelB, a suppressor of canonical NF-κB, and exhibited altered cellular localization of this protein. Our results indicate that MALT1 is a central, cell-intrinsic factor that determines the encephalitogenic potential of inflammatory Th17 cells in vivo.
Article
Full-text available
Inflammation conveys the development of glomerular injury and is a major cause of progressive kidney disease. NF-κB signaling is among the most important regulators of pro-inflammatory signaling. Its role in podocytes, the epithelial cells at the kidney filtration barrier, is poorly understood. Here, we inhibited NF-κB signaling in podocytes by specific ablation of the NF-κB essential modulator (NEMO, IKKγ). Podocyte-specific NEMO-deficient mice (NEMO(pko)) were viable and did not show proteinuria or overt changes in kidney morphology. After induction of glomerulonephritis both NEMO(pko) and control mice developed significant proteinuria. However, NEMO(pko) mice recovered much faster, showing rapid remission of proteinuria and restoration of podocyte morphology. Interestingly, quantification of infiltrating macrophages, T-lymphocytes and granulocytes at day 7 revealed no significant difference between wild-type and NEMO(pko). To further investigate the underlying mechanisms we created a stable NEMO knock-down mouse podocyte cell line. Again, no overt changes in morphology were observed. Translocation of NF-κB to the nucleus after stimulation with TNFα or IL-1was sufficiently inhibited. Moreover, secretion of proinflammatory chemokines from podocytes after stimulation with TNFα or IL-1 was significantly reduced in NEMO deficient podocytes and in glomerular samples obtained at day 7 after induction of nephrotoxic nephritis. Collectively, these results show that proinflammatory activity of NF-κB in podocytes aggravates proteinuria in experimental glomerulonephritis in mice. Based on these data it may be speculated that immunosuppressive drugs may not only target professional immune cells but also podocytes directly to convey their beneficial effects in various types of glomerulonephritis.
Article
Migratory non-lymphoid tissue dendritic cells (NLT-DCs) transport antigens to lymph nodes (LNs) and are required for protective immune responses in the context of inflammation and to promote tolerance to self-antigens in steady-state. However, the molecular mechanisms that elicit steady-state NLT-DC maturation and migration are unknown. By comparing the transcriptome of NLT-DCs in the skin with their migratory counterparts in draining LNs, we have identified a novel NF-κB-regulated gene network specific to migratory DCs. We show that targeted deletion of IKKβ in DCs, a major activator of NF-κB, prevents NLT-DC accumulation in LNs and compromises regulatory T cell conversion in vivo. This was associated with impaired tolerance and autoimmunity. NF-κB is generally considered the prototypical pro-inflammatory transcription factor, but this study describes a role for NF-κB signaling in DCs for immune homeostasis and tolerance that could have implications in autoimmune diseases and immunity. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Immune responses in the skin are important for host defence against pathogenic microorganisms. However, dysregulated immune reactions can cause chronic inflammatory skin diseases. Extensive crosstalk between the different cellular and microbial components of the skin regulates local immune responses to ensure efficient host defence, to maintain and restore homeostasis, and to prevent chronic disease. In this Review, we discuss recent findings that highlight the complex regulatory networks that control skin immunity, and we provide new paradigms for the mechanisms that regulate skin immune responses in host defence and in chronic inflammation.
Article
The NF-kappa B signal transduction pathway is best known as a major regulator of innate and adaptive immune responses, yet there is a growing appreciation of its importance in immune cell development, particularly of T lineage cells. In this Review, we discuss how the temporal regulation of NF-kappa B controls the stepwise differentiation and antigen-dependent selection of conventional and specialized subsets of T cells in response to T cell receptor and costimulatory, cytokine and growth factor signals.
Article
The nuclear factor-κB (NF-κB) family of transcription factors plays important roles in various biological processes including apoptosis, stress response, immunity, and inflammation. NF-κB signaling is involved in both immune cell development and function, and it is critical in modulation of the immune response through the transcriptional regulation of cytokine and chemokine expression. An area of great interest in T-cell-mediated adaptive immunity is the ability of naive CD4(+) T cells generated in the thymus to differentiate into various subsets including T-helper 1 (Th1), Th2, Th17, Th9, follicular helper T (Tfh), Th22, and regulatory T (Treg) cells, upon encountering different pathogens and microenvironments. In this review, we discuss the role of NF-κB pathway in the development and functional divergence of the different helper T-cell subsets as well as in regulatory T cells.