SlideShare a Scribd company logo
1 of 11
Download to read offline
Enzymatic autoantibody glycan hydrolysis alleviates autoimmunity against type
VII collagen
Misa Hirose a
, Katerina Vafia a
, Kathrin Kalies b
, Stephanie Groth a
, Jürgen Westermann b
, Detlef Zillikens a
,
Ralf J. Ludwig a
, Mattias Collin d
, Enno Schmidt a,c,*
a
Department of Dermatology, University of Lübeck, Ratzeburger Allee 160, Lübeck D-23538, Germany
b
Institute of Anatomy, University of Lübeck, Ratzeburger Allee 160, Lübeck D-23538, Germany
c
Comprehensive Center for Inflammation Medicine, University of Lübeck, Ratzeburger Allee 160, Lübeck D-23538, Germany
d
Division of Infection Medicine, Department of Clinical Science, Lund University, SE-22184 Lund, Sweden
a r t i c l e i n f o
Article history:
Received 11 December 2011
Received in revised form
4 April 2012
Accepted 12 April 2012
Keywords:
Autoantibody
Bullous
EndoS
IgG
Hydrolysis
a b s t r a c t
Autoantibody-mediated diseases comprise a heterogeneous group of disorders in which the pathogenic
potential of autoantibodies has been clearly demonstrated. In general, their treatment relies on the long-
term use of systemic corticosteroids and other immunosuppressants that are associated with consid-
erable adverse reactions. EndoS, an endoglycosidase derived from Streptococcus pyogenes, specifically
hydrolyzes the N-linked glycan of native IgG and has previously been shown to modulate the interaction
between the Fc portion of autoantibody and Fcg receptors on leukocytes. Here, different models of
autoimmunity to type VII collagen, a structural protein of the dermal-epidermal junction (DEJ), were
employed to explore the therapeutic potential of EndoS. First, pretreatment of otherwise pathogenic
anti-murine type VII collagen (mCOL7) IgG with EndoS significantly reduced split formation at the DEJ in
cryosections of murine skin and abrogated clinical disease in mice. Next, the effect of EndoS was also
seen when the enzyme was injected into mice after pathogenic anti-mCOL7 IgG had been administered.
Finally, to mimic the patient situation even closer, EndoS was applied in mice that had already developed
clinical disease after immunization with mCOL7. In all EndoS-treated mice, disease progression was
stopped, and in the majority of mice, clinical disease even regressed. Of note, EndoS was shown to
hydrolyze already in vivo-bound pathogenic autoantibodies. In addition, EndoS treatment decreased
lesional expression of activating FcgRs while increasing FcgRIIB expression.
Ó 2012 Elsevier Ltd.
1. Introduction
Autoantibody-mediated autoimmune diseases comprise
a heterogeneous group of disorders in which the pathogenic rele-
vance of autoantibodies has been demonstrated, e.g. immune-
mediated thrombocytopenia, ANCA-mediated vasculitis, primary
progressive multiple sclerosis, Sjögren’s syndrome, anti-
phospholipid syndrome, systemic lupus erythematosus, rheuma-
toid arthritis as well as pemphigus and pemphigoid diseases [1,2].
Pemphigus and pemphigoid diseases are characterized by autoan-
tibodies against desmosomal and hemidesmosomal antigens,
respectively [3e5]. The hemidesmosomal proteins collagen type VII
and XVII have been identified as target antigens of epidermolysis
bullosa acquisita (EBA) and bullous pemphigoid, respectively [6e9].
The role of autoantibodies against collagen type VII and XVII has
clearly been established [10e14].
Type VII collagen is a major component of anchoring fibrils in
the basal membrane zone of the skin and mucous membranes with
squamous epithelia [15,16]. Epitopes recognized by the majority of
EBA sera were mapped to the non-collagenous (NC) 1 domain of
type VII collagen [17e19]. We and others have shown the patho-
genic relevance of anti-type VII collagen antibodies by several
in vitro and in vivo models. Specifically, anti-type VII collagen
antibodies from EBA patients induced dermal-epidermal separa-
tion in cryosections of human skin [20], rabbit and patients type VII
collagen-specific IgG reproduced the clinical and immunopatho-
logical features of the human disease when passively transferred
into mice [11,21,22], and immunization of certain mouse strains
with a recombinant fragment of the murine NC1 domain of type VII
collagen resulted in a blistering phenotype closely resembling
* Corresponding author. Department of Dermatology, University of Lübeck, Rat-
zeburger Allee 160, D-23538 Lübeck, Germany. Tel.: þ49 (0)451 500 2538; fax: þ49
(0)451 500 2981.
E-mail address: enno.schmidt@uk-sh.de (E. Schmidt).
Contents lists available at SciVerse ScienceDirect
Journal of Autoimmunity
journal homepage: www.elsevier.com/locate/jautimm
0896-8411 Ó 2012 Elsevier Ltd.
doi:10.1016/j.jaut.2012.04.002
Journal of Autoimmunity 39 (2012) 304e314
Open access under CC BY-NC-ND license.
Open access under CC BY-NC-ND license.
human EBA [23]. In these experimental models, binding of anti-
bodies to its target antigen alone was not sufficient for lesion
formation but complement activation, the recruitment and acti-
vation of leukocytes, and the release of proteases, that finally led to
the dermal-epidermal separation, were essential [24e26]. The
central role of the Fc portion of anti-type VII collagen IgG was
shown by the lack of pathogenicity of both rabbit F(ab’)2 fragments
and chicken IgY against anti-type VII collagen [11,27].
The glycosylation status of the Fc portion of IgG is important for
interaction with Fcg receptors (FcgRs). In fact, hydrolysis of IgG
glycan reduces the binding ability to FcgRs in both humans and
mice [28,29]. EndoS, derived from Streptococcus pyogenes, is an
endoglycosidase that hydrolyzes the conserved N-linked glycan on
IgG heavy chains [30e32]. To date, EndoS is the only known
endoglycosidase that exclusively hydrolyzes the glycan of native
IgG [32,33].
Treatment of most autoantibody-mediated diseases, including
EBA, is largely unspecific and relies on the long-term use of
systemic corticosteroids and additional immunosuppressants that
is frequently associated with severe adverse reactions. Thus, novel
therapeutic avenues are warranted and indeed, EndoS has previ-
ously been successfully applied in experimental autoimmune
diseases, including rheumatoid arthritis [34] and immune-
mediated thrombocytopenia [32]. However, the effect of EndoS
on autoantibodies that have already bound to their target antigens
and that have already induced clinical disease has not yet been
addressed.
Here, we showed that EndoS significantly reduced the patho-
genic effect of autoantibodies to type VII collagen in 3 different
experimental models. Importantly, closely mimicking the patient
situation, EndoS suppressed disease activity in the immunization-
induced mouse model, even when applied after anti- collagen
type VII antibodies had bound to the skin and clinical disease had
developed. Extending the present knowledge of the action of EndoS
the enzyme was shown to hydrolyze already in vivo-bound path-
ogenic autoantibodies and to differentially modulate the expres-
sion of activating and inhibiting FcgRs.
2. Materials and methods
2.1. Generation of rabbit antibodies to murine collagen type VII
(mCOL7)
Pathogenic IgG was generated as previously described [11]. In
brief, New Zealand white rabbits were immunized with recombi-
nant forms of the glutathione S-transferase (GST)-tagged NC1
domain of mCOL7 and the rabbit serum was purified by affinity
chromatography using protein G. Reactivity of IgG fractions was
analyzed by indirect immunofluorescence (IF) microscopy on
murine (BALB/c) skin. In parallel, rabbit IgG against GST alone was
prepared.
2.2. EndoS preparation and IgG hydrolysis in vitro
Recombinant GST-tagged EndoS was expressed in Escherichia
coli as previously described [33]. One mg of rabbit anti-mCOL7 IgG
was incubated with 5 mg recombinant GST-EndoS in PBS for 16 h at
37 C followed by affinity removal of GST-EndoS by serial passages
over Glutathione-Sepharose 4B columns (GE Healthcare, Uppsala,
Sweden). This GST-EndoS-treated IgG is called EndoS-pretreated
IgG throughout the paper. Prior treatment of animals, the GST-tag
was removed from GST-EndoS using factor Xa (Novagen, Madi-
son, WI). Liberated GST and factor Xa was removed by serial passage
over Xarrest agarose (Novagen) and Glutathione-Sepharose 4B.
2.3. IgG purification, SDS-PAGE, and lectin blotting
For sodium dodecyl sulphate polyacrylamide gel electrophoresis
(SDS-PAGE) and lectin blot analyses, total IgG was purified from
10 ml mouse serum samples using Ab SpinTrap (GE Healthcare)
according to manufacturer’s instructions. SDS-PAGE and immuno-
blotting were performed as previously described [32]. Glycosylated
IgG was detected by using 5 mg/ml biotinylated Lens culinaris
agglutinin (LCA)-lectin (Vector Laboratories, Burlingame, CA) and
1 mg/ml streptavidinehorseradish peroxidase (Vector Laboratories)
and Super Signal West Pico peroxidase substrate (Pierce, Rockford,
IL). Membranes were analyzed by using a Chemidoc XRS imaging
system and Quantity One image analysis software (BioRad,
Hercules, CA).
2.4. Cryosection assay
Skin dermal-epidermal separation was evaluated using an
ex vivo model as previously described [35]. Briefly, leukocytes from
the peripheral blood of healthy volunteers were isolated by a sedi-
mentation gradient containing sodium diatrizoate and dextran 500
following the instructions of the manufacturer (Nycomed, Oslo,
Norway), then harvested and washed twice in RPMI 1640 (Life
Technologies, Karlsruhe, Germany) and resuspended in the same
medium. The cell suspension was kept on ice and cell viability was
tested using trypan blue; preparations with viability greater than
95% were used. Cryosections prepared from adult mouse tail skin
(BALB/c) were placed in the center of a Superfrost Plus microscope
slide (Menzel-Glaser, Braunschweig, Germany). Skin sections were
washed with PBS for 5 min to remove embedding medium, then
incubated with 50 ml of 1:2 diluted either EndoS-pretreated anti-
mCOL7 IgG or non-treated anti-mCOL7 IgG for 90 min at room
temperature. After washing the sections with PBS twice, chambers
were prepared as described and the leukocyte suspension at the
concentration of 1  107
cell/ml was injected into the chambers.
Subsequently, chambers were disassembled, and sections were
washed in PBS, fixed in formalin, and stained with hematoxylin and
eosin. Finally, skin dermal-epidermal separation was evaluated
microscopically and the extent expressed as the proportion of the
epidermis detached from the dermis. Approval for these studies
was obtained from the Institutional Review board at the University
of Lübeck (Lübeck, Germany 09-140), and informed consent was
provided according to the Declaration of Helsinki.
2.5. Induction of autoimmunity to mCOL7
2.5.1. Animals
Six to 8 week-old BALB/c mice for passive EBA model and SJL for
active EBA model were purchased from Charles River (Sulzfeld,
Germany). Animals were housed at 12 h lightedark cycle at the
experimental animal facility in the University of Lübeck. All clinical
examinations, biopsies and bleedings were performed under
anesthesia using intraperitoneal administration of a mixture of
ketamine (100 mg/g) and xylazine (15 mg/g). Animal experiments
were approved by local authorities of the Animal Care and Use
Committee (Kiel, Germany; 68-6/09) and performed by certified
personnel.
2.5.2. Passive transfer model
Passive transfer studies followed a previously published
protocol [11] with minor modifications. Briefly, mice received
7.5 mg of rabbit anti-mCOL7 IgG or EndoS-pretreated rabbit anti-
mCOL7 IgG subcutaneously in abdominal skin every other day
from day 0 in total of 6 injections. Alternatively, EndoS was injected
i.p. 24 h after anti-mCOL7 IgG injections. Mice were evaluated at 4
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 305
day-intervals for presence of skin lesions until 12 days after first IgG
injection (day 12). Cutaneous lesions were counted, and the extent
of skin disease was scored as involvement of the skin surface as
reported previously [11]. At the same time intervals, blood was
obtained for analysis of circulating antibodies. At day 12, all mice
were sacrificed, blood was taken, and both lesional and perilesional
biopsies were taken for histopathological analysis (stored in 4%
buffered formalin) and perilesional biopsies were direct IF
microscopy for determination of IgG and C3 deposits at the DEJ
(stored at 80 C), respectively.
2.5.3. Immunization-induced model
Immunization-induced autoimmunity to mCOL7 was generated
as previously reported [36,37]. In brief, SJL mice were immunized at
the hind footpad with 60 mg of recombinant murine type VII
collagen immunodominant peptide emulsified in the non-ionic
block copolymer adjuvant TiterMax (ALEXIS Biochemicals, Lör-
rach, Germany). When mice had developed disease with a clinical
score greater 2, they were randomly allocated into 3 experimental
groups (week 0). Two groups were injected intraperitoneally with
200 mg EndoS and 400 mg EndoS, respectively, once a week for 4
weeks. Mice were clinically evaluated at a week intervals for
presence of skin lesions until week 4; at the same time blood was
taken. Cutaneous lesions were counted, and the extent of skin
disease was scored as involvement of the skin surface as reported
previously [23]. At the end of week 4, biopsies were taken and
blood was drawn as described above for the passive transfer model.
Circulating autoantibodies against mCOL7 were detected by ELISA
as previously described [37].
2.6. Histopathology
Formalin-fixed skin samples were processed into paraffin
blocks. 4 mm sections were stained with hematoxylin and eosin
according to standard protocols [37].
2.7. Immunohistochemistry
Six mm cryosections of lesional skin obtained from mice were
fixed in acetone at 20 C for 10 min. Air dried sections were
washed with PBS for 5 min 3 times and incubated with Dako Dual
Endogenous Enzyme Block (DakoCytomation, Hamburg, Germany)
Fig. 1. EndoS-treated pathogenic IgG did not induce separation at the dermal-epidermal junction (DEJ) of normal mouse skin ex vivo. Six mm cryosections incubated with non-
treated (a) and EndoS-pretreated (b) rabbit anti-mCOL7 IgG. After addition of granulocytes from healthy volunteers for a 1 h incubation time, significantly more neutrophils
were recruited along the DEJ in sections incubated with untreated compared to sections incubated with EndoS-treated rabbit anti-mCOL7 IgG (c; ***p  0.001, t-test). White
triangles (D) indicate the DEJ. After an incubation period of 3 h, significantly less subepidermal split formation was seen in sections incubated with untreated rabbit anti-mCOL7 IgG
(d) compared to sections incubated with EndoS-treated rabbit anti-mCOL7 IgG (e). The size of separation at the DEJ is significantly less in EndoS-pretreated rabbit anti-mCOL7 IgG
incubated sections compared with non-treated rabbit anti-mCOL7 IgG (f). *p  0.05 (t-test). The experiment was conducted six times in triplicate for the evaluation of the DEJ
separation. For the neutrophil binding evaluation, one experiment in triplicate was performed. The extent of neutrophil binding at the DEJ of the two random areas of the entire
section with high power view (400) and of DEJ separation was evaluated by two blinded observers.
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314
306
for 30 min at room temperature (RT) for blocking endogenous
peroxidase. To reduce non-specific binding of secondary antibody,
sections were incubated with 5% normal rabbit serum for 20 min at
RT. After excess serum from sections was blotted, sections were
incubated with rat anti-mouse Ly-6C/ly-6G (Gr-1; RB56-8C5, BD
Pharmingen, San Diego, CA, USA) antibody (1:200 dilution in 2%
normal rabbit serum/PBS) for 1 h at RT. Slides were washed with
PBS followed by incubation with rabbit anti-rat IgG (1:200 dilution
in 2% normal rabbit serum/PBS; DakoCytomation) for 45 min at RT.
After washing, sections were incubated with R.T.U. VECTASTAINÒ
Elite ABC Reagent (Vector Laboratories) for 30 min at RT. To develop
the peroxidase reaction, sections were incubated in HistoGreen
(LINARIS, Wertheim-Bettingen, Germany) solution until appro-
priate color developed. Excess solution was washed in tap water,
followed by counterstaining with hematoxylin and sections were
mounted.
2.8. Immunofluorescence (IF) microscopy
Direct IF microscopy to detect tissue-bound IgG, complement C3
deposits has been described previously [23]. Staining intensity of
immunoreactants at the DEJ was quantified with ImageJ software
(http://rsbweb.nih.gov/ij/). Six mm sections of healthy BALB/c
mouse skin were prepared for indirect IF microscopy to evaluate
tissue binding ability of rabbit anti-mCOL7 IgG and mouse sera [37].
Binding of autoantibodies and C5 to the skin was visualized using
FITC-conjugated rabbit anti-mouse IgG and the monoclonal anti-
mouse C5 antibody BB5.1, respectively (Dako and Cell Sciences,
Canton, MA, USA).
2.9. Quantitive reverse transcriptase-polymerase chain reaction
(RT-PCR)
To evaluate the FcgR and inflammatory cytokine expression in
the skin, total RNA was isolated from the half of the right ear
according to the manufacturer’s protocol (Analytic Jena Ag, Neu
Ruppin, Germany). The RNA was incubated with DNase I
(SigmaeAldrich Chemie, Munich, Germany) for 15 min and fol-
lowed by heating at 70 C for 10 min, then immediately cooled on
ice. After reverse transcription the cDNA was added to the qPCR
Master Mix Plus SYBR Green I (Eurogentec, Cologne, Germany) and
amplified using the SDS ABI 7900 system (Applied Biosystems,
Darmstadt, Germany). The forward and reverse primes were
designed by using the computer software CloneManager (Sci Ed
Central, Version7.01; Cary, NC, USA). The optimal primmer
concentrations used were 500 nM each for the forward and reverse
primers (biomers.net, Ulm, Germany). Primers used for this project
are shown in Supplementary Table 1. The same batch of cDNA
(20 ml) was used to determine the cycles of threshold (ct), and the
amount of the cytokine cDNA copies were normalized to the house
keeping gene MLN51.
2.10. Autoantibody elution from the skin
Mice (n ¼ 4) were injected twice with rabbit anti-mCOL7 IgG at
day 0 and 2. On day 4, two mice were injected with EndoS 400 mg
and the other two mice remained untreated. Twenty-four hours
later (day 5) mice were sacrificed and skin was sampled and
at 80 C. Ten mm sections were prepared and placed on Superfrost
Plus microscope slides (Menzel-Glaeser). After removing bedding
medium by washing with PBS, antibodies were eluted from the skin
washing with 0.1M glycin, pH 2.8 at 4 C as described previously
[38]. The collected antibodies were neutralized with TriseHCl and
concentrated (Millipore, Cork, Ireland and Vivaspin 500, Sartorius
Stedim, Goettingen, Germany). Autoantibody binding was verified
by indirect IF microscopy on mouse skin.
2.11. Statistical analysis
Sigma plot 11.0 (Systat Software, Inc, Chicago, USA) was used to
perform statistical analysis. A p-value of 0.05 was considered
statistically significant. Applied tests are indicated at the respective
table or figure legends.
cl
l
∗∗
∗∗∗ ∗∗∗
b
c
a
r
Fig. 2. EndoS-pretreatment of anti-mCOL7 IgG failed to induce clinical disease in vivo. BALB/c mice were subcutaneously injected with 7.5 mg per head EndoS-treated (n ¼ 8) and
untreated rabbit anti-mCOL7 IgG (n ¼ 11) every other day over 12 days (total 45 mg). Clinical scores were evaluated every 4 days and determined as the extent of body surface
affected by lesions. Mice injected with EndoS-pretreated rabbit anti-mCOL7 IgG demonstrate a significantly lower clinical score compared with mice that received untreated rabbit
anti-mCOL7 IgG alone (a; ***p  0.001, **p  0.01, t-test). Representative clinical pictures of mice injected with untreated rabbit mCOL7 IgG (b) or EndoS-pretreated rabbit anti-
mCOL7 IgG (c) at day 12.
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 307
3. Results
3.1. Hydrolyzed anti-mouse type VII collagen IgG did not change the
binding capacity to its target antigen in vitro
The IgG glycan hydrolyzing effect of EndoS treatment was
verified by SDS-PAGE and Lens culinaris agglutinin (LCA) lectin blot
analysis before EndoS-treated IgG was applied in further experi-
ments. After successful IgG glycan hydrolysis, SDS-PAGE and LCA
lectin blot analysis showed a reduction of the molecular size of the
heavy chain of rabbit anti-mouse type VII collagen (mCOL7) IgG by
approximately 3 kDa and a strongly reduced signal of the EndoS-
treated rabbit anti-mCOL7 IgG compared to untreated IgG,
respectively (Supplementary Fig. 1). Serial dilutions of EndoS-
treated and untreated anti-mCOL7 IgG used in equal concentra-
tions resulted in the same titer (1:51,200) and binding pattern by
indirect immunofluorescence (IF) microscopy on mouse skin (data
not shown). Thus, hydrolyzation by EndoS did not affect the F(ab’)2
fragment function, i.e. the binding of anti-anti-mCOL7 IgG to the
dermal-epidermal junction (DEJ).
3.2. EndoS-pretreatment of otherwise pathogenic IgG failed to
induce dermal-epidermal separation ex vivo
To evaluate the effect of EndoS treatment, cryosections of
normal mouse skin were incubated in vitro with EndoS-treated and
untreated rabbit anti-mCOL7 IgG, respectively. After addition of
granulocytes from healthy volunteers for a 1 h incubation time,
significantly more neutrophils were recruited along the DEJ in
sections incubated with untreated compared to sections incubated
with EndoS-treated anti-mCOL7 IgG (Fig. 1aec; p  0.001). After an
incubation period of 3 h, significantly less subepidermal split
formation was observed in sections incubated with EndoS-treated
IgG compared to sections incubated with untreated anti-mCOL7
IgG (Fig. 1def; p  0.05).
3.3. EndoS-pretreatment of otherwise pathogenic IgG failed to
induce clinical disease in vivo
Since the ex vivo pathogenicity of rabbit anti-mCOL7 IgG was
abrogated after treatment with EndoS, the next set of experiences
addressed the in vivo effect of EndoS treatment. BALB/c mice were
subcutaneously injected with 7.5 mg per head EndoS-treated (n ¼ 8)
and untreated anti-mCOL7 IgG (n ¼ 11) every other day over 12 days.
Mice injected with EndoS-treated IgG (Fig. 2c) did not develop
clinical disease while, as expected, mice injected with untreated IgG
(Fig. 2b) developed severe skin lesion reproducing the human
disease (Fig. 2a). These data were obtained from 2 independent
experiments with different anti-mCOL7 IgG preparations.
3.4. Treatment of EndoS following injection of pathogenic IgG
suppressed disease severity in vivo
To evaluate the effect of EndoS in vivo, BALB/c mice received
intraperitoneal EndoS every other day, starting at day 1 until day 11,
at a dose of 10 mg/injection (n ¼ 8) or 100 mg/injection (n ¼ 8) 24 h
after anti-mCOL7 IgG injection. Disease severity was significantly
milder in mice that had received 100 mg EndoS in conjunction with
anti-mCOL7 IgG compared to both mice that had been injected with
10 mg EndoS plus anti-mCOL7 IgG and mice that had received anti-
mCOL7 IgG alone (Fig. 3a). The IgG glycan status of mouse serum
was analyzed in all mice at day 12. SDS-PAGE and LCA lectin blot-
ting confirmed that IgG from all EndoS-treated mice was signifi-
cantly less glycosylated compared to IgG from mice that had not
been treated with EndoS (Fig. 3b).
3.5. Autoantibody-mediated intralesional inflammatory responses
were inhibited by EndoS
The inflammatory response in skin lesions of mice was subse-
quently analyzed by histopathology and quantification of mRNA for
different cytokines, chemokines, and cellular markers, including
∗∗
a
b
cl
l
r
Fig. 3. Treatment of EndoS following injection of pathogenic IgG suppressed disease
severity in vivo. EndoS was applied at a dose of 10 mg/injection (n ¼ 8) or 100 mg/
injection (n ¼ 8), respectively, every other day, starting at day 1 until day 11, 24 h after
rabbit anti-mCOL7 IgG injection. Disease severity was significantly milder in mice that
had received 100 mg EndoS in conjunction with rabbit anti-mCOL7 IgG compared to
mice that had received rabbit anti-mCOL7 IgG alone. (a; **p  0.01, One-way ANOVA).
The IgG glycan status of serum was analyzed in all mice obtained at day 12. SDS-PAGE
(Stain) and Lens culinaris agglutinin (LCA) lectin blotting confirmed that in the 100 mg
EndoS group and, to a lesser extent, in the 10 mg Endo group, IgG was considerably less
hydrolyzed compared to IgG from untreated mice. Results form 8 individual mice of
each group are shown. Blots were performed in the same experiment including the
appropriate controls and were rearranged for better comparison (b).
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314
308
FcgR, by RT-PCR. By histopathology of biopsies from mouse ears,
significantly less inflammatory cells were observed in the dermis of
both mice that had received EndoS-pretreated anti-mCOL7 IgG
(n ¼ 8) and mice had been injected with EndoS along with anti-
mCOL7 IgG (n ¼ 8) compared to mice that were injected with
anti-mCOL7 IgG alone (n ¼ 11; Fig. 4aec). Also, mRNA levels of T cell
(CD3), dendritic cell (CD11c), macrophage and monocyte markers
(Mac-1) as well as of chemokines (KC, MIP) and inflammatory
cytokines (TNFa, and IL-1b) were significantly lower in skin of mice
that had received EndoS-pretreated anti-mCOL7 IgG (n ¼ 8)
compared to mice that had obtained untreated anti-mCOL7 IgG
(n ¼ 11; p  0.001 for CD3, KC, MIP, and IL-1b, p  0.05 for CD11c,
p  0.01 for TNFa; Fig. 4d). In contrast, mRNA levels of IFN-g,
RANTES, and IL-6 remained unaltered (p ¼ 0.570, p ¼ 0.483 and
p ¼ 0.06, respectively).
3.6. Autoantibody-mediated in vivo deposition of IgG and
complement was unaltered by EndoS
In order to investigate the effect of EndoS on the binding of
autoantibodies to their target antigen and subsequent complement
recruitment, in vivo deposition of IgG, C3, and C5 at the DEJ were
visualized by direct IF microscopy of mouse ear skin obtained from
the three experimental groups: anti-mCOL7 IgG alone (Fig. 5aec),
EndoS-pretreated anti-mCOL7 IgG (Fig. 5def), and EndoS in
conjunction with anti-mCOL7 IgG (gei). The intensity of fluores-
cence was quantified using ImageJ software. The levels of tissue-
bound IgG (Fig. 5j), C3 (Fig. 5k), and C5 (Fig. 5l) were not signifi-
cantly different between the three experimental group. These data
indicate the EndoS did not interfere with autoantibody binding and
complement recruitment in vivo.
3.7. Therapeutic efficacy of EndoS in immunization-induced
autoimmunity
To mimic the therapeutic situation in patients with
autoantibody-mediated disorders more closely, mice (n ¼ 50) were
immunized with a recombinant portion of the immunodominant
NC1 domain of murine type VII collagen. Mice that developed
a disease score of greater 2 (between 4 and 8 weeks after immu-
nization) were allocated to three groups: no treatment (n ¼ 16), i.p.
injection of 200 mg (n ¼ 11) and 400 mg (n ¼ 12) EndoS once per
week for 4 weeks, respectively. Mice in both EndoS treatment
groups developed significantly less disease compared to non-
treated mice (Fig. 6aec). Importantly, disease progression was
already abrogated after the first treatment week in the high-dose
group pointing to a direct effect of EndoS on the glycosylation
status of in vivo-bound autoantibody. In addition, the number of
mice in which disease activity decreased below the clinical score at
the time of treatment allocation was significantly higher in the
EndoS groups (11 of 23) compared to the untreated mice (0 of 16;
p ¼ 0.00065, Fisher’s exact test). Histopathological analysis showed
significantly less infiltration of inflammatory cells including Gr-1
positive cells in dermal lesions of EndoS-treated mice compared
Fig. 4. Autoantibody-mediated intralesional inflammatory responses were inhibited by EndoS. Evaluation of the inflammatory response in ear skin was performed by histopa-
thology (aec) and quantification of mRNA for different chemokines, cytokines, and cell markers by RT-PCR (d). Representative histopathology pictures of skin obtained from mice
injected with untreated anti-mCOL7 IgG alone demonstrates significantly higher inflammatory cell infiltration (a) compared with skin from mice injected with EndoS-pretreated
anti-mCOL7 IgG (b) or untreated anti-mCOL7 IgG together with 100 mg of EndoS (c). By RT-PCR, mRNA levels of makers for T cells (CD3), dendritic cell (CD11c) and macrophages/
monocytes (Mac-1) as well as of chemokines (KC and MIP) and inflammatory cytokines (TNFa and IL-1b) were significantly less in skin of mice that received EndoS-pretreated anti-
mCOL7 IgG (n ¼ 8) compared to that of untreated anti-mCOL7 IgG-injected mice (n ¼ 11; d; *p  0.05, **p  0.01, ***p  0.001, t-test).
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 309
to untreated animals (Fig. 6fei). By direct IF microscopy, IgG and C3
deposits at the DEJ did not differ between any of the groups
(Fig. 6jem).
3.8. EndoS does hydrolyze already-tissue-bound autoantibodies
Since EndoS injection reversed already established clinical
disease we further evaluated if EndoS could hydrolyze autoanti-
bodies that had already bound to their target antigen in the tissue.
IgG that was eluted from the skin following binding of pathogenic
anti-mCOL7 IgG and subsequent EndoS treatment showed size
reduction by SDS-PAGE and was unreactive by lectin blotting in
contrast to IgG eluted from the skin of mice that did not receive
EndoS (Fig. 7). Both eluted IgG fractions equally labeled the DEJ of
mouse skin by indirect IF microscopy (Fig. 7).
3.9. EndoS modulates the expression of Fcg receptors in
autoantibody-mediated skin destruction
In mice that received EndoS-pretreated anti-mCOL7 IgG, mRNA
levels of the activating FcgRs were significantly reduced compared
to mice injected with untreated anti-mCOL7 IgG (Fig. 8a). In
Fig. 5. Autoantibody-mediated in vivo deposition of IgG and complement was unaltered by EndoS. By direct immunofluorescence microscopy of non-lesional mouse skin obtained
from mice injected with anti-COL7 IgG (a, b, c), EndoS-pretreated anti-COL7 IgG (d, e, f), and anti-mCOL7 IgG together with 100 mg of EndoS (g, h, i), in vivo deposits of IgG (a, d, g),
C3 (b, e, h) and C5 (c, f, i) at the dermal-epidermal junction were visualized. The intensity of fluorescence was quantified using ImageJ software (j, IgG; k, C3; l, C5). No significant
difference was observed between the 3 groups.
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314
310
contrast, no differences were seen in mRNA levels of the inhibitory
FcgRIIB between the two groups (Fig. 8a). Similar data were ob-
tained when FcgR expression was related to the CD11c expression
in lesional skin, however, statistical significance was not reached
(Fig. 8b). When FcgR expression was analyzed in the immunization-
induced model, EndoS injections resulted in reduced expression of
the activating FcgR and increased expression of FcgRIIB (Fig. 8c).
When related to the lesional expression of CD11c, a statistical
significant downregulation was observed for FcgRIII and FcgRIV
whereas expression of FcgRIIB was significantly increased (Fig. 8d).
4. Discussion
EndoS has been identified as glycosidase that specifically
hydrolyzes the conserved N-linked glycan on IgG heavy chains
[30e32]. EndoS has a unique specificity for native IgG or IgG Fc
portion glycans compared to related endoglycosidases which are
specific for certain glycans that can be attached to any protein or
peptide, and which are enhanced by or require denaturation of the
protein backbone [32,33]. By hydrolysis of IgG glycan, binding
affinity of the Fc portion of IgG to Fcg receptors (FcgRs) is modu-
lated. More precisely, EndoS-treated IgG has a considerably lower
affinity to FcgRI, FcgRIIB, FcgRIII, and FcgRIV in mice [29]. Inter-
estingly, EndoS-hydrolyzed human IgG2 binds better to human
inhibitory receptor FcRIIB, while binding of all EndoS-hydrolyzed
IgG subclasses to activating receptors is lowered [28]. The thera-
peutic efficacy of EndoS has previously been demonstrated in
different experimental autoimmune diseases induced by passive
transfer of antibodies, including rheumatoid arthritis [29,34],
immune-mediated thrombocytopenia [32], hemolytic anemia [39],
and ANCA-mediated glomerulonephritis [40]. Since EndoS does not
specifically target pathogenic autoantibodies but hydrolyzes the
entire pool of IgG, it may increase the risk for infections and
interfere with vaccination-induce immunity in human patients. On
the other hand, EndoS does not affect IgG’s F(ab)’ binding capacity
and activation of the classical complement system [32] leaving
intact large parts of the humoral immune defense system.
Furthermore, repetitive injections with EndoS can raise EndoS-
specific antibodies which may hamper its efficacy.
Here, we extended the current knowledge about the mecha-
nisms of action of EndoS by the use of an immunization-induced
mouse model in which EndoS exerted its effect on autoantibodies
that had already bound to their autoantigen. In addition, expression
of the activating FcgRIII and FcgRIV in the skin lesions was down-
regulated whereas expression of the inhibiting FcgRIIB was upre-
gulated following EndoS treatment.
In the first set of experiments, pretreatment of pathogenic
anti-mCOL7 IgG with EndoS resulted in a statistically significant
reduction of dermal-epidermal separation in cryosections of mouse
Fig. 6. Therapeutic efficacy of EndoS in immunization-induced autoimmunity. Mice
that developed a disease score of greater 2 (between 4 and 8 weeks after
immunization with a recombinant portion of the immunodominant NC1 domain of
murine type VII collagen) were allocated into three groups: no treatment (n ¼ 16) and
i.p. injection of 200 mg (n ¼ 11) or 400 mg (n ¼ 12) EndoS once per week for 4 weeks,
respectively. Mice in both EndoS treatment groups developed significantly less disease
compared to non-treated mice (a; *p  0.05). Representative clinical (bee), histo-
pathological (f,g,h,i), and immunopathological pictures (jem) of untreated mice (b, f, h,
j, l) and 400 mg EndoS-treated mice (c, e, g, i, k, m) are shown. Ear of a mouse that did
not receive EndoS (b) demonstrates more clinical disease, e.g. erythema and erosion,
compared with that was treated with 100 mg EndoS for 4 weeks (c). Clinical lesion on
a mouse tail at week 0 (before treatment, d) was completely healed after 4 weeks of
400 mg EndoS treatment (e). Histopathology of 400 mg EndoS-treated mice (g) shows
significantly less infiltration of inflammatory cells including Gr-1 positive cells (gran-
ulocytes and monocytes) demonstrated by immunohistochemistry (i) in dermal
lesions compared with those of untreated mice (f, h). By direct IF microscopy, IgG (j, k)
and C3 (l, m) deposits at the DEJ did not differ between non-treated (j, l) and EndoS-
treated (k, m) groups.
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 311
skin. In this ex vivo model, cryosections of mouse skin were incu-
bated with anti-mCOL7 IgG and subsequently with leukocytes from
healthy volunteers. Leukocytes attached to the DEJ and via the
release of elastase and matrix metalloproteinase-9 from gran-
ulocytes, the DEJ is split [26]. Based on these findings, in the next
set of experiments, a mouse model was used in which the injection
of anti-mCOL7 IgG reproduces the clinical and immunopathological
characteristics of the human disease [11]. In this model, the
pretreatment of pathogenic anti-mCOL7 IgG with EndoS prevented
clinical disease in all mice. When EndoS was administrated to mice
24 h after the injection of pathogenic anti-mCOL7 IgG, disease
activity was significantly reduced. In order to control for a potential
effect of EndoS on the interaction of anti-mCOL7 IgG with mCOL7,
Fig. 7. EndoS hydrolyzes tissue-bound autoantibodies IgG eluted from the skin of mice
that had received anti-mCOL7 IgG followed by injection of EndoS (þ) showed size
reduction by Coomassie Brilliant Blue staining (Stain) and lost reactivity by blotting
with LCA lectin (LCA blot). In comparison, IgG eluted from the skin of mice that were
injected with anti-mCOL7 IgG alone is shown (). Both eluted IgG fraction labeled the
dermal-epidermal junction by indirect immunofluorescence microscopy on normal
murine skin (IF microscopy).
a b
c
d
Fig. 8. EndoS differently modulates the expression of activating and inhibitory Fcg receptors mRNA expression of Fc gamma receptors (FcgRs) in the lesional skin was determined.
Expression of activating FcgRs (FcgRI, FcgRIII, and FcgRIV) in EndoS-pretreated anti-mCOL7 IgG-injected mice was significantly less compared with untreated pathogenic IgG-
injected animals (a; *p  0.05, t-test). Expression of FcgRIIB was significantly increased following EndoS treatment (a; *p  0.05, t-test). Similar data were obtained in the
immunization-induced mouse model (c). Here, data reached statistical significance only for FcgRIV (c; *p  0.05, t-test). When the lesional FcgR expression was related to the
expression of CD11c in both experimental models, a decrease of the activating and increase of the inhibitory FcgR(s) was seen (b, d). Statistical significance was observed in the
immunization-induced model (d; *p  0.05, t-test).
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314
312
the binding of anti-mCOL7 IgG to mouse skin was visualized by
direct IF microscopy. No significant difference between skin of mice
that received anti-mCOL7 IgG alone, EndoS-treated anti-mCOL7
IgG, and pathogenic IgG, followed by EndoS injections, was
observed.
Furthermore, the binding of C3 and C5 to the DEJ remained
unaltered between the 3 groups. Previously, the binding of C3 and
C5 to the DEJ and the activation of the alternative pathway were
shown to be critical for disease development in this model [24].
EndoS has previously been shown to prevent binding of C1q,
a component of the classical pathway, to IgG-sensitized red blood
cells and subsequent hemolysis [39]. These data suggest that EndoS
may interfere with the binding of IgG to components of the clas-
sical, but not of the alternative pathway of complement activation.
In autoimmunity to mCOL7, complement deposition at the DEJ is
thought to be an essential step to attract inflammatory cells that
then become activated via the interaction between the Fc portion of
autoantibody and FcgR, which further enhances the local inflam-
mation, and finally leads to the release of proteases at the DEJ.
Regarding the pathogenic role of complement, we observed
unchanged levels of tissue-bound C3 and C5. It has been proposed
that anaphylatoxin C5a, which does not bind to tissue, and C5aR on
neutrophils may compose an amplification loop for neutrophil
activation in ANCA-mediated nephritis model [41]. This speculates
that the less neutrophils due to inhibited the binding of FcgRs on
neutrophils and Fc part of the pathogenic IgG caused less C5a
production, and eventually reduce the disease severity at the lesion.
Our results indicate that EndoS treatment interferes with this
further enhancement of the local inflammation, thus inhibiting
dermal-epidermal separation.
To mimic the situation of EBA patients more closely, in the last set
of experiments, an immunization-induced model was employed. In
this model, SJL mice develop the clinical and immunopathological
hallmarks of the human disease 4e8 weeks after immunizationwith
a recombinant fragment of the immunodominant NC1 domain of
mCOL7 [23]. This mouse model appeared to be particularly attrac-
tive since development of clinical disease depended on the depo-
sition of IgG2b autoantibodies to the DEJ [23] and EndoS was shown
to only functionally impair IgG1 and IgG2b subclasses in mice [34].
In the present study, after disease onset, mice were allocated to 3
groups: 2 groups received weekly injections of different EndoS
doses and the third group was left untreated. In both treatment
groups, progression of the disease was abrogated. In fact, clinical
disease was reduced in nearly half of EndoS-treated mice but in none
of untreated animals.
Importantly, in the high-dose EndoS group, progression of the
clinical disease activity was already inhibited one week after the
first EndoS injection. In the low-dose EndoS group, this effect was
observed one week later. Since anti-mCOL7 IgG bound in murine
skin was previously found to remain in place for about 8 weeks by
investigating the off-spring of mice immunized with recombinant
mCOL7 [36], the clinical effect of EndoS in the present study is most
likely explained by a functional interaction of EndoS with tissue-
bound anti-mCOL7 IgG. This assumption was experimentally
corroborated when skin-bound anti-mCOL7 IgG eluted after in vivo
EndoS treatment was shown to be hydrolyzed in contrast to eluted
anti-mCOL7 IgG without preceding EndoS treatment. These find-
ings have implications on a future therapeutic use of EndoS in
autoantibody-mediated diseases in which EndoS may interfere not
only with serum autoantibodies but also with autoantibodies that
have already bound to their target tissue.
Finally, we explored the effect of EndoS on the composition of
the inflammatory infiltrate and the differential expression of FcgR
in skin. We found mRNA levels of the different FcgR in the mouse
skin not to be equally affected by EndoS treatment, which has not
previously been reported. In both the passive transfer model and
immunization-induced model, the activating FcgRs, including
FcgRI and low affinity FcgRIII and FcgRIV were significantly
downregulated, while the inhibiting FcgRIIB was upregulated
following EndoS treatment. This effect was shown to be in part
attributed to the reduction of the inflammatory infiltrate in the skin
lesions. In addition, a novel independent effect of EndoS on the
FcgR expression was observed. The pathogenic relevance of upre-
gulated activating FcgRs and downregulated FcgRIIB has previously
been shown in autoimmunity including human and experimental
rheumatoid arthritis, experimental arthritis and immune-complex
mediated nephritis [42,43] as well as, most recently, autoimmunity
to type VII collagen [44].
In conclusion, we extended the present knowledge about the
mechanisms of action of EndoS and favor the further exploration of
enzymatic autoantibody glycan hydrolysis as novel, therapeutic
option in severe autoantibody-mediated disease.
Conflicts of interest disclosure
Patents for the in vitro and in vivo use of EndoS have been
applied for by Genovis AB and Hansa Medical AB, respectively. M.C.
is listed as an inventor on these applications that are pending.
Hansa Medical AB in part funded this study, but had no influence on
the design of study, interpretation of data, or the final form of the
manuscript. M.C. is a part time scientific consultant for Hansa
Medical AB.
Acknowledgments
Ulla Johannesson, Lund, and Rebecca Cames and Claudia Kau-
derer, Lübeck, are acknowledged for excellent technical assistance.
This work was supported by grants from the Excellence Cluster
“Inflammation at Interfaces” (EXC306/1), Novartis Pharma (to E.S.)
as well as the Swedish Research Council (projects 2005-4791 and
2010-57X-20240), the Foundations of Crafoord, Hedberg, Bergvall,
Österlund, Wiberg, Söderberg, the Swedish Society for Medicine,
the Royal Physiografic Society, King Gustaf V’s Memorial Fund, the
Swedish Rheumatism Association, the Medical Faculty at Lund
University, and Hansa Medical AB to M.C.
Appendix A. Supplementary material
Supplementary data related to this article can be found, in the
online version at, doi:10.1016/j.jaut.2012.04.002.
References
[1] Lim PL, Zouali M. Pathogenic autoantibodies: emerging insights into tissue
injury. Immunol Lett 2006;103:17e26.
[2] Nandakumar KS, Holmdahl R. Therapeutic cleavage of IgG: new avenues for
treating inflammation. Trends Immunol 2008;29:173e8.
[3] Stanley JR, Amagai M. Pemphigus, bullous impetigo, and the staphylococcal
scalded-skin syndrome. N Engl J Med 2006;355:1800e10.
[4] Schmidt E, Zillikens D. Modern diagnosis of autoimmune blistering skin
diseases. Autoimmun Rev 2010;10:84e9.
[5] Sticherling M, Erfurt-Berge C. Autoimmune blistering diseases of the skin.
Autoimmun Rev; 2011.
[6] Woodley DT, Briggaman RA, O’Keefe EJ, Inman AO, Queen LL, Gammon WR.
Identification of the skin basement-membrane autoantigen in epidermolysis
bullosa acquisita. N Engl J Med 1984;310:1007e13.
[7] Woodley DT, Burgeson RE, Lunstrum G, Bruckner-Tuderman L, Reese MJ,
Briggaman RA. Epidermolysis bullosa acquisita antigen is the globular
carboxyl terminus of type VII procollagen. J Clin Invest 1988;81:683e7.
[8] Labib RS, Anhalt GJ, Patel HP, Mutasim DF, Diaz LA. Molecular heterogeneity of
the bullous pemphigoid antigens as detected by immunoblotting. J Immunol
1986;136:1231e5.
[9] Giudice GJ, Emery DJ, Diaz LA. Cloning and primary structural analysis of the
bullous pemphigoid autoantigen BP180. J Invest Dermatol 1992;99:243e50.
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 313
[10] Chen M, Keene DR, Costa FK, Tahk SH, Woodley DT. The carboxyl terminus of
type VII collagen mediates antiparallel dimer formation and constitutes a new
antigenic epitope for epidermolysis bullosa acquisita autoantibodies. J Biol
Chem 2001;276:21649e55.
[11] Sitaru C, Mihai S, Otto C, Chiriac MT, Hausser I, Dotterweich B, et al. Induction
of dermal-epidermal separation in mice by passive transfer of antibodies
specific to type VII collagen. J Clin Invest 2005;115:870e8.
[12] Liu Z, Diaz LA, Troy JL, Taylor AF, Emery DJ, Fairley JA, et al. A passive transfer
model of the organ-specific autoimmune disease, bullous pemphigoid, using
antibodies generated against the hemidesmosomal antigen, BP180. J Clin
Invest 1993;92:2480e8.
[13] Nishie W, Sawamura D, Goto M, Ito K, Shibaki A, McMillan JR, et al. Human-
ization of autoantigen. Nat Med 2007;13:378e83.
[14] Recke A, Sitaru C, Vidarsson G, Evensen M, Chiriac MT, Ludwig RJ, et al.
Pathogenicity of IgG subclass autoantibodies to type VII collagen: induction of
dermal-epidermal separation. J Autoimmun 2010;34:435e44.
[15] Sakai LY, Keene DR, Morris NP, Burgeson RE. Type VII collagen is a major
structural component of anchoring fibrils. J Cell Biol 1986;103:1577e86.
[16] Wetzels RH, Robben HC, Leigh IM, Schaafsma HE, Vooijs GP, Ramaekers FC.
Distribution patterns of type VII collagen in normal and malignant human
tissues. Am J Pathol 1991;139:451e9.
[17] Lapiere JC, Woodley DT, Parente MG, Iwasaki T, Wynn KC, Christiano AM, et al.
Epitope mapping of type VII collagen. Identification of discrete peptide
sequences recognized by sera from patients with acquired epidermolysis
bullosa. J Clin Invest 1993;92:1831e9.
[18] Gammon WR, Murrell DF, Jenison MW, Padilla KM, Prisayanh PS, Jones DA,
et al. Autoantibodies to type VII collagen recognize epitopes in a fibronectin-
like region of the noncollagenous (NC1) domain. J Invest Dermatol 1993;100:
618e22.
[19] Tanaka T, Furukawa F, Imamura S. Epitope mapping for epidermolysis bullosa
acquisita autoantibody by molecularly cloned cDNA for type VII collagen.
J Invest Dermatol 1994;102:706e9.
[20] Sitaru C, Kromminga A, Hashimoto T, Brocker EB, Zillikens D. Autoantibodies
to type VII collagen mediate Fcgamma-dependent neutrophil activation and
induce dermal-epidermal separation in cryosections of human skin. Am J
Pathol 2002;161:301e11.
[21] Woodley DT, Chang C, Saadat P, Ram R, Liu Z, Chen M. Evidence that anti-type
VII collagen antibodies are pathogenic and responsible for the clinical,
histological, and immunological features of epidermolysis bullosa acquisita.
J Invest Dermatol 2005;124:958e64.
[22] Woodley DT, Ram R, Doostan A, Bandyopadhyay P, Huang Y, Remington J,
et al. Induction of epidermolysis bullosa acquisita in mice by passive transfer
of autoantibodies from patients. J Invest Dermatol 2006;126:1323e30.
[23] Sitaru C, Chiriac MT, Mihai S, Buning J, Gebert A, Ishiko A, et al. Induction of
complement-fixing autoantibodies against type VII collagen results in
subepidermal blistering in mice. J Immunol 2006;177:3461e8.
[24] Mihai S, Chiriac MT, Takahashi K, Thurman JM, Holers VM, Zillikens D, et al.
The alternative pathway of complement activation is critical for blister
induction in experimental epidermolysis bullosa acquisita. J Immunol 2007;
178:6514e21.
[25] Chiriac MT, Roesler J, Sindrilaru A, Scharffetter-Kochanek K, Zillikens D,
Sitaru C. NADPH oxidase is required for neutrophil-dependent autoantibody-
induced tissue damage. J Pathol 2007;212:56e65.
[26] Shimanovich I, Mihai S, Oostingh GJ, Ilenchuk TT, Brocker EB, Opdenakker G,
et al. Granulocyte-derived elastase and gelatinase B are required for dermal-
epidermal separation induced by autoantibodies from patients with epi-
dermolysis bullosa acquisita and bullous pemphigoid. J Pathol 2004;204:
519e27.
[27] Sesarman A, Mihai S, Chiriac MT, Olaru F, Sitaru AG, Thurman JM, et al. Binding
of avian IgY to type VII collagen does not activate complement and leucocytes
and fails to induce subepidermal blistering in mice. Br J Dermatol 2008;158:
463e71.
[28] Allhorn M, Olin AI, Nimmerjahn F, Collin M. Human IgG/Fc gamma R inter-
actions are modulated by streptococcal IgG glycan hydrolysis. PLoS One 2008;
3:e1413.
[29] Nandakumar KS, Collin M, Olsen A, Nimmerjahn F, Blom AM, Ravetch JV, et al.
Endoglycosidase treatment abrogates IgG arthritogenicity: importance of IgG
glycosylation in arthritis. Eur J Immunol 2007;37:2973e82.
[30] Collin M, Olsen A. EndoS, a novel secreted protein from Streptococcus pyo-
genes with endoglycosidase activity on human IgG. EMBO J 2001;20:
3046e55.
[31] Collin M, Svensson MD, Sjoholm AG, Jensenius JC, Sjobring U, Olsen A. EndoS
and SpeB from Streptococcus pyogenes inhibit immunoglobulin-mediated
opsonophagocytosis. Infect Immun 2002;70:6646e51.
[32] Collin M, Shannon O, Bjorck L. IgG glycan hydrolysis by a bacterial enzyme as
a therapy against autoimmune conditions. Proc Natl Acad Sci U S A 2008;105:
4265e70.
[33] Collin M, Olsen A. Effect of SpeB and EndoS from Streptococcus pyogenes on
human immunoglobulins. Infect Immun 2001;69:7187e9.
[34] Albert H, Collin M, Dudziak D, Ravetch JV, Nimmerjahn F. In vivo enzymatic
modulation of IgG glycosylation inhibits autoimmune disease in an IgG
subclass-dependent manner. Proc Natl Acad Sci U S A 2008;105:15005e9.
[35] Sitaru C, Schmidt E, Petermann S, Munteanu LS, Brocker EB, Zillikens D. Auto-
antibodies to bullous pemphigoid antigen 180 induce dermal-epidermal
separation in cryosections of human skin. J Invest Dermatol 2002;118:664e71.
[36] Kasperkiewicz M, Hirose M, Recke A, Schmidt E, Zillikens D, Ludwig RJ.
Clearance rates of circulating and tissue-bound autoantibodies to type VII
collagen in experimental epidermolysis bullosa acquisita. Br J Dermatol 2010;
162:1064e70.
[37] Ludwig RJ, Recke A, Bieber K, Muller S, Marques Ade C, Banczyk D, et al.
Generation of antibodies of distinct subclasses and specificity is linked to H2s
in an active mouse model of epidermolysis bullosa acquisita. J Invest Dermatol
2011;131:167e76.
[38] Korman NJ. In situ-bound antibodies eluted from the skin of patients with
bullous pemphigoid are preferentially directed against the 230-kD bullous
pemphigoid antigen. J Invest Dermatol 1995;105:824e30.
[39] Allhorn M, Briceno JG, Baudino L, Lood C, Olsson ML, Izui S, et al. The IgG-
specific endoglycosidase EndoS inhibits both cellular and complement-
mediated autoimmune hemolysis. Blood 2010;115:5080e8.
[40] van Timmeren MM, van der Veen BS, Stegeman CA, Petersen AH, Hellmark T,
Collin M, et al. IgG glycan hydrolysis attenuates ANCA-mediated glomerulo-
nephritis. J Am Soc Nephrol 2010;21:1103e14.
[41] Schreiber A, Xiao H, Jennette JC, Schneider W, Luft FC, Kettritz R. C5a receptor
mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am
Soc Nephrol 2009;20:289e98.
[42] Wijngaarden S, van de Winkel JG, Jacobs KM, Bijlsma JW, Lafeber FP, van
Roon JA. A shift in the balance of inhibitory and activating Fcgamma receptors
on monocytes toward the inhibitory Fcgamma receptor IIb is associated with
prevention of monocyte activation in rheumatoid arthritis. Arthritis Rheum
2004;50:3878e87.
[43] Nimmerjahn F, Lux A, Albert H, Woigk M, Lehmann C, Dudziak D, et al.
FcgammaRIV deletion reveals its central role for IgG2a and IgG2b activity
in vivo. Proc Natl Acad Sci U S A 2010;107:19396e401.
[44] Kasperkiewicz M, Nimmerjahn F, Wende S, Hirose M, Iwata H, Jonkman MF,
et al. Genetic identification and functional validation of FcgammaRIV as key
molecule in autoantibody-induced tissue injury. J Pathol; 2012.
M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314
314

More Related Content

Similar to Enzymatic-autoantibody-glycan-hydrolysis-alleviates-auto_2012_Journal-of-Aut.pdf

Monoclonal antibodies
Monoclonal antibodiesMonoclonal antibodies
Monoclonal antibodiesSupriyaDaphal
 
vaccins drug delivery system
vaccins drug delivery systemvaccins drug delivery system
vaccins drug delivery systemManish Jajodiya
 
New approaches for medical management of Chronic Granulomatous Disease.
New approaches for medical management of Chronic Granulomatous Disease.New approaches for medical management of Chronic Granulomatous Disease.
New approaches for medical management of Chronic Granulomatous Disease.Dmitri Popov
 
Infections in immunocompromised patients
Infections in immunocompromised patientsInfections in immunocompromised patients
Infections in immunocompromised patientsجهاد الخريصي
 
Rabies Virus_Presentation_by_Maliha_Rashid.pptx
Rabies Virus_Presentation_by_Maliha_Rashid.pptxRabies Virus_Presentation_by_Maliha_Rashid.pptx
Rabies Virus_Presentation_by_Maliha_Rashid.pptxMaliha Rashid
 
Format 2016: masqueradesyndromes in allergicdiseases.
Format 2016: masqueradesyndromes in allergicdiseases.Format 2016: masqueradesyndromes in allergicdiseases.
Format 2016: masqueradesyndromes in allergicdiseases.Envicon Medical Srl
 
Advances_in_immunology_in_aibd.pptx
Advances_in_immunology_in_aibd.pptxAdvances_in_immunology_in_aibd.pptx
Advances_in_immunology_in_aibd.pptxHIMABINDUMAMIDALA2
 
Controversy: the role of immunomodulator in allergic case
Controversy: the role of immunomodulator in allergic caseControversy: the role of immunomodulator in allergic case
Controversy: the role of immunomodulator in allergic caseSuharti Wairagya
 
BIOTEM: Covid-19 and Serological Tests
BIOTEM: Covid-19 and Serological Tests BIOTEM: Covid-19 and Serological Tests
BIOTEM: Covid-19 and Serological Tests Jonathan Mayali
 
Marios_Stylianou_PhD thesis
Marios_Stylianou_PhD thesisMarios_Stylianou_PhD thesis
Marios_Stylianou_PhD thesisMarios Stylianou
 
Immunity and its applied aspect
Immunity and its applied aspectImmunity and its applied aspect
Immunity and its applied aspectDrRoopse Singh
 
SHILPA COMMON.pptx
SHILPA COMMON.pptxSHILPA COMMON.pptx
SHILPA COMMON.pptxmalti19
 

Similar to Enzymatic-autoantibody-glycan-hydrolysis-alleviates-auto_2012_Journal-of-Aut.pdf (20)

Monoclonal antibodies
Monoclonal antibodiesMonoclonal antibodies
Monoclonal antibodies
 
vaccins drug delivery system
vaccins drug delivery systemvaccins drug delivery system
vaccins drug delivery system
 
Lupus
LupusLupus
Lupus
 
New approaches for medical management of Chronic Granulomatous Disease.
New approaches for medical management of Chronic Granulomatous Disease.New approaches for medical management of Chronic Granulomatous Disease.
New approaches for medical management of Chronic Granulomatous Disease.
 
Pediatr allergy immunol 2009 20 266 272
Pediatr allergy immunol 2009 20 266 272Pediatr allergy immunol 2009 20 266 272
Pediatr allergy immunol 2009 20 266 272
 
Th2 cytokines
Th2 cytokines Th2 cytokines
Th2 cytokines
 
Infections in immunocompromised patients
Infections in immunocompromised patientsInfections in immunocompromised patients
Infections in immunocompromised patients
 
Intravenous immunoglobulin (IVIG)
Intravenous immunoglobulin (IVIG)Intravenous immunoglobulin (IVIG)
Intravenous immunoglobulin (IVIG)
 
mAb
mAbmAb
mAb
 
Rabies Virus_Presentation_by_Maliha_Rashid.pptx
Rabies Virus_Presentation_by_Maliha_Rashid.pptxRabies Virus_Presentation_by_Maliha_Rashid.pptx
Rabies Virus_Presentation_by_Maliha_Rashid.pptx
 
Format 2016: masqueradesyndromes in allergicdiseases.
Format 2016: masqueradesyndromes in allergicdiseases.Format 2016: masqueradesyndromes in allergicdiseases.
Format 2016: masqueradesyndromes in allergicdiseases.
 
Advances_in_immunology_in_aibd.pptx
Advances_in_immunology_in_aibd.pptxAdvances_in_immunology_in_aibd.pptx
Advances_in_immunology_in_aibd.pptx
 
Controversy: the role of immunomodulator in allergic case
Controversy: the role of immunomodulator in allergic caseControversy: the role of immunomodulator in allergic case
Controversy: the role of immunomodulator in allergic case
 
BIOTEM: Covid-19 and Serological Tests
BIOTEM: Covid-19 and Serological Tests BIOTEM: Covid-19 and Serological Tests
BIOTEM: Covid-19 and Serological Tests
 
Marios_Stylianou_PhD thesis
Marios_Stylianou_PhD thesisMarios_Stylianou_PhD thesis
Marios_Stylianou_PhD thesis
 
Knockout mouse for allergy
Knockout mouse for allergyKnockout mouse for allergy
Knockout mouse for allergy
 
Zoonotic Viral Disease.pptx
Zoonotic Viral Disease.pptxZoonotic Viral Disease.pptx
Zoonotic Viral Disease.pptx
 
Immunity and its applied aspect
Immunity and its applied aspectImmunity and its applied aspect
Immunity and its applied aspect
 
SHILPA COMMON.pptx
SHILPA COMMON.pptxSHILPA COMMON.pptx
SHILPA COMMON.pptx
 
Hepatit konusmasi-klimik-2013
Hepatit konusmasi-klimik-2013Hepatit konusmasi-klimik-2013
Hepatit konusmasi-klimik-2013
 

More from melymar1978

ijms-20-02494.pdf
ijms-20-02494.pdfijms-20-02494.pdf
ijms-20-02494.pdfmelymar1978
 
INSTRUMENTOS ODONTOLÓGICOS.pdf
INSTRUMENTOS ODONTOLÓGICOS.pdfINSTRUMENTOS ODONTOLÓGICOS.pdf
INSTRUMENTOS ODONTOLÓGICOS.pdfmelymar1978
 
1-s2.0-S0896841112000455-main.pdf
1-s2.0-S0896841112000455-main.pdf1-s2.0-S0896841112000455-main.pdf
1-s2.0-S0896841112000455-main.pdfmelymar1978
 
RIUNNE_AR_Britos_MR.pdf
RIUNNE_AR_Britos_MR.pdfRIUNNE_AR_Britos_MR.pdf
RIUNNE_AR_Britos_MR.pdfmelymar1978
 
Copia de Online Notebook Pink variant _ by Slidesgo_.pdf
Copia de Online Notebook Pink variant _ by Slidesgo_.pdfCopia de Online Notebook Pink variant _ by Slidesgo_.pdf
Copia de Online Notebook Pink variant _ by Slidesgo_.pdfmelymar1978
 
1-s2.0-S0718539112700903-main.pdf
1-s2.0-S0718539112700903-main.pdf1-s2.0-S0718539112700903-main.pdf
1-s2.0-S0718539112700903-main.pdfmelymar1978
 
1-s2.0-S0718539113701319-main.pdf
1-s2.0-S0718539113701319-main.pdf1-s2.0-S0718539113701319-main.pdf
1-s2.0-S0718539113701319-main.pdfmelymar1978
 
EXAMEN PRÁCTICO I UNIDAD.pdf
EXAMEN PRÁCTICO I UNIDAD.pdfEXAMEN PRÁCTICO I UNIDAD.pdf
EXAMEN PRÁCTICO I UNIDAD.pdfmelymar1978
 
Caso clínico.pdf
Caso clínico.pdfCaso clínico.pdf
Caso clínico.pdfmelymar1978
 
EXAMEN PRÁCTICO I UNIDAD (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1).pdfEXAMEN PRÁCTICO I UNIDAD (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1).pdfmelymar1978
 
CASO CLÍNICO I UNIDAD (1).pdf
CASO CLÍNICO I UNIDAD (1).pdfCASO CLÍNICO I UNIDAD (1).pdf
CASO CLÍNICO I UNIDAD (1).pdfmelymar1978
 
CASO CLÍNICO I UNIDAD.pdf
CASO CLÍNICO I UNIDAD.pdfCASO CLÍNICO I UNIDAD.pdf
CASO CLÍNICO I UNIDAD.pdfmelymar1978
 
EXAMEN PRÁCTICO I UNIDAD (1) (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1) (1).pdfEXAMEN PRÁCTICO I UNIDAD (1) (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1) (1).pdfmelymar1978
 

More from melymar1978 (16)

ijms-20-02494.pdf
ijms-20-02494.pdfijms-20-02494.pdf
ijms-20-02494.pdf
 
INSTRUMENTOS ODONTOLÓGICOS.pdf
INSTRUMENTOS ODONTOLÓGICOS.pdfINSTRUMENTOS ODONTOLÓGICOS.pdf
INSTRUMENTOS ODONTOLÓGICOS.pdf
 
1-s2.0-S0896841112000455-main.pdf
1-s2.0-S0896841112000455-main.pdf1-s2.0-S0896841112000455-main.pdf
1-s2.0-S0896841112000455-main.pdf
 
RIUNNE_AR_Britos_MR.pdf
RIUNNE_AR_Britos_MR.pdfRIUNNE_AR_Britos_MR.pdf
RIUNNE_AR_Britos_MR.pdf
 
10371.pdf
10371.pdf10371.pdf
10371.pdf
 
Copia de Online Notebook Pink variant _ by Slidesgo_.pdf
Copia de Online Notebook Pink variant _ by Slidesgo_.pdfCopia de Online Notebook Pink variant _ by Slidesgo_.pdf
Copia de Online Notebook Pink variant _ by Slidesgo_.pdf
 
art02.pdf
art02.pdfart02.pdf
art02.pdf
 
1-s2.0-S0718539112700903-main.pdf
1-s2.0-S0718539112700903-main.pdf1-s2.0-S0718539112700903-main.pdf
1-s2.0-S0718539112700903-main.pdf
 
v26n1a05.pdf
v26n1a05.pdfv26n1a05.pdf
v26n1a05.pdf
 
1-s2.0-S0718539113701319-main.pdf
1-s2.0-S0718539113701319-main.pdf1-s2.0-S0718539113701319-main.pdf
1-s2.0-S0718539113701319-main.pdf
 
EXAMEN PRÁCTICO I UNIDAD.pdf
EXAMEN PRÁCTICO I UNIDAD.pdfEXAMEN PRÁCTICO I UNIDAD.pdf
EXAMEN PRÁCTICO I UNIDAD.pdf
 
Caso clínico.pdf
Caso clínico.pdfCaso clínico.pdf
Caso clínico.pdf
 
EXAMEN PRÁCTICO I UNIDAD (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1).pdfEXAMEN PRÁCTICO I UNIDAD (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1).pdf
 
CASO CLÍNICO I UNIDAD (1).pdf
CASO CLÍNICO I UNIDAD (1).pdfCASO CLÍNICO I UNIDAD (1).pdf
CASO CLÍNICO I UNIDAD (1).pdf
 
CASO CLÍNICO I UNIDAD.pdf
CASO CLÍNICO I UNIDAD.pdfCASO CLÍNICO I UNIDAD.pdf
CASO CLÍNICO I UNIDAD.pdf
 
EXAMEN PRÁCTICO I UNIDAD (1) (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1) (1).pdfEXAMEN PRÁCTICO I UNIDAD (1) (1).pdf
EXAMEN PRÁCTICO I UNIDAD (1) (1).pdf
 

Recently uploaded

Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...
Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...
Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...Pooja Nehwal
 
Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...
Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...
Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...amitlee9823
 
(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escortsranjana rawat
 
Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...
Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...
Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...amitlee9823
 
9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...
9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...
9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...Pooja Nehwal
 
Call Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance BookingCall Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance Bookingroncy bisnoi
 
Call Girls Dubai Slut Wife O525547819 Call Girls Dubai Gaped
Call Girls Dubai Slut Wife O525547819 Call Girls Dubai GapedCall Girls Dubai Slut Wife O525547819 Call Girls Dubai Gaped
Call Girls Dubai Slut Wife O525547819 Call Girls Dubai Gapedkojalkojal131
 
Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...
Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...
Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...Pooja Nehwal
 
Deira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort Girls
Deira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort GirlsDeira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort Girls
Deira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort GirlsEscorts Call Girls
 
Call Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur High Profile
 
VIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 BookingVIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 Bookingdharasingh5698
 
Call Girls Kothrud Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Kothrud Call Me 7737669865 Budget Friendly No Advance BookingCall Girls Kothrud Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Kothrud Call Me 7737669865 Budget Friendly No Advance Bookingroncy bisnoi
 
(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)
(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)
(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)kojalkojal131
 
Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...
Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...
Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...ranjana rawat
 
Top Rated Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...
Top Rated  Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...Top Rated  Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...
Top Rated Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...Call Girls in Nagpur High Profile
 
Lubrication and it's types and properties of the libricabt
Lubrication and it's types and properties of the libricabtLubrication and it's types and properties of the libricabt
Lubrication and it's types and properties of the libricabtdineshkumar430venkat
 
Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...
Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...
Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...Naicy mandal
 

Recently uploaded (20)

Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...
Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...
Call Girls in Thane 9892124323, Vashi cAll girls Serivces Juhu Escorts, powai...
 
young call girls in Sainik Farm 🔝 9953056974 🔝 Delhi escort Service
young call girls in Sainik Farm 🔝 9953056974 🔝 Delhi escort Serviceyoung call girls in Sainik Farm 🔝 9953056974 🔝 Delhi escort Service
young call girls in Sainik Farm 🔝 9953056974 🔝 Delhi escort Service
 
Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...
Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...
Call Girls Banashankari Just Call 👗 7737669865 👗 Top Class Call Girl Service ...
 
(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
(PARI) Alandi Call Girls Just Call 7001035870 [ Cash on Delivery ] Pune Escorts
 
Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...
Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...
Vip Mumbai Call Girls Andheri East Call On 9920725232 With Body to body massa...
 
CHEAP Call Girls in Mayapuri (-DELHI )🔝 9953056974🔝(=)/CALL GIRLS SERVICE
CHEAP Call Girls in Mayapuri  (-DELHI )🔝 9953056974🔝(=)/CALL GIRLS SERVICECHEAP Call Girls in Mayapuri  (-DELHI )🔝 9953056974🔝(=)/CALL GIRLS SERVICE
CHEAP Call Girls in Mayapuri (-DELHI )🔝 9953056974🔝(=)/CALL GIRLS SERVICE
 
9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...
9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...
9892124323, Call Girl in Juhu Call Girls Services (Rate ₹8.5K) 24×7 with Hote...
 
Call Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance BookingCall Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Pimple Saudagar Call Me 7737669865 Budget Friendly No Advance Booking
 
Call Girls Dubai Slut Wife O525547819 Call Girls Dubai Gaped
Call Girls Dubai Slut Wife O525547819 Call Girls Dubai GapedCall Girls Dubai Slut Wife O525547819 Call Girls Dubai Gaped
Call Girls Dubai Slut Wife O525547819 Call Girls Dubai Gaped
 
Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...
Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...
Kalyan callg Girls, { 07738631006 } || Call Girl In Kalyan Women Seeking Men ...
 
Deira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort Girls
Deira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort GirlsDeira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort Girls
Deira Dubai Escorts +0561951007 Escort Service in Dubai by Dubai Escort Girls
 
Call Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Sakshi Call 7001035870 Meet With Nagpur Escorts
 
VIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 BookingVIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 Booking
VIP Call Girls Dharwad 7001035870 Whatsapp Number, 24/07 Booking
 
Call Girls Kothrud Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Kothrud Call Me 7737669865 Budget Friendly No Advance BookingCall Girls Kothrud Call Me 7737669865 Budget Friendly No Advance Booking
Call Girls Kothrud Call Me 7737669865 Budget Friendly No Advance Booking
 
(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)
(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)
(=Towel) Dubai Call Girls O525547819 Call Girls In Dubai (Fav0r)
 
Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...
Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...
Book Paid Lohegaon Call Girls Pune 8250192130Low Budget Full Independent High...
 
Top Rated Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...
Top Rated  Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...Top Rated  Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...
Top Rated Pune Call Girls Katraj ⟟ 6297143586 ⟟ Call Me For Genuine Sex Serv...
 
Lubrication and it's types and properties of the libricabt
Lubrication and it's types and properties of the libricabtLubrication and it's types and properties of the libricabt
Lubrication and it's types and properties of the libricabt
 
Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...
Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...
Makarba ( Call Girls ) Ahmedabad ✔ 6297143586 ✔ Hot Model With Sexy Bhabi Rea...
 
Vip Call Girls Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS Live
Vip Call Girls Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS LiveVip Call Girls Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS Live
Vip Call Girls Noida ➡️ Delhi ➡️ 9999965857 No Advance 24HRS Live
 

Enzymatic-autoantibody-glycan-hydrolysis-alleviates-auto_2012_Journal-of-Aut.pdf

  • 1. Enzymatic autoantibody glycan hydrolysis alleviates autoimmunity against type VII collagen Misa Hirose a , Katerina Vafia a , Kathrin Kalies b , Stephanie Groth a , Jürgen Westermann b , Detlef Zillikens a , Ralf J. Ludwig a , Mattias Collin d , Enno Schmidt a,c,* a Department of Dermatology, University of Lübeck, Ratzeburger Allee 160, Lübeck D-23538, Germany b Institute of Anatomy, University of Lübeck, Ratzeburger Allee 160, Lübeck D-23538, Germany c Comprehensive Center for Inflammation Medicine, University of Lübeck, Ratzeburger Allee 160, Lübeck D-23538, Germany d Division of Infection Medicine, Department of Clinical Science, Lund University, SE-22184 Lund, Sweden a r t i c l e i n f o Article history: Received 11 December 2011 Received in revised form 4 April 2012 Accepted 12 April 2012 Keywords: Autoantibody Bullous EndoS IgG Hydrolysis a b s t r a c t Autoantibody-mediated diseases comprise a heterogeneous group of disorders in which the pathogenic potential of autoantibodies has been clearly demonstrated. In general, their treatment relies on the long- term use of systemic corticosteroids and other immunosuppressants that are associated with consid- erable adverse reactions. EndoS, an endoglycosidase derived from Streptococcus pyogenes, specifically hydrolyzes the N-linked glycan of native IgG and has previously been shown to modulate the interaction between the Fc portion of autoantibody and Fcg receptors on leukocytes. Here, different models of autoimmunity to type VII collagen, a structural protein of the dermal-epidermal junction (DEJ), were employed to explore the therapeutic potential of EndoS. First, pretreatment of otherwise pathogenic anti-murine type VII collagen (mCOL7) IgG with EndoS significantly reduced split formation at the DEJ in cryosections of murine skin and abrogated clinical disease in mice. Next, the effect of EndoS was also seen when the enzyme was injected into mice after pathogenic anti-mCOL7 IgG had been administered. Finally, to mimic the patient situation even closer, EndoS was applied in mice that had already developed clinical disease after immunization with mCOL7. In all EndoS-treated mice, disease progression was stopped, and in the majority of mice, clinical disease even regressed. Of note, EndoS was shown to hydrolyze already in vivo-bound pathogenic autoantibodies. In addition, EndoS treatment decreased lesional expression of activating FcgRs while increasing FcgRIIB expression. Ó 2012 Elsevier Ltd. 1. Introduction Autoantibody-mediated autoimmune diseases comprise a heterogeneous group of disorders in which the pathogenic rele- vance of autoantibodies has been demonstrated, e.g. immune- mediated thrombocytopenia, ANCA-mediated vasculitis, primary progressive multiple sclerosis, Sjögren’s syndrome, anti- phospholipid syndrome, systemic lupus erythematosus, rheuma- toid arthritis as well as pemphigus and pemphigoid diseases [1,2]. Pemphigus and pemphigoid diseases are characterized by autoan- tibodies against desmosomal and hemidesmosomal antigens, respectively [3e5]. The hemidesmosomal proteins collagen type VII and XVII have been identified as target antigens of epidermolysis bullosa acquisita (EBA) and bullous pemphigoid, respectively [6e9]. The role of autoantibodies against collagen type VII and XVII has clearly been established [10e14]. Type VII collagen is a major component of anchoring fibrils in the basal membrane zone of the skin and mucous membranes with squamous epithelia [15,16]. Epitopes recognized by the majority of EBA sera were mapped to the non-collagenous (NC) 1 domain of type VII collagen [17e19]. We and others have shown the patho- genic relevance of anti-type VII collagen antibodies by several in vitro and in vivo models. Specifically, anti-type VII collagen antibodies from EBA patients induced dermal-epidermal separa- tion in cryosections of human skin [20], rabbit and patients type VII collagen-specific IgG reproduced the clinical and immunopatho- logical features of the human disease when passively transferred into mice [11,21,22], and immunization of certain mouse strains with a recombinant fragment of the murine NC1 domain of type VII collagen resulted in a blistering phenotype closely resembling * Corresponding author. Department of Dermatology, University of Lübeck, Rat- zeburger Allee 160, D-23538 Lübeck, Germany. Tel.: þ49 (0)451 500 2538; fax: þ49 (0)451 500 2981. E-mail address: enno.schmidt@uk-sh.de (E. Schmidt). Contents lists available at SciVerse ScienceDirect Journal of Autoimmunity journal homepage: www.elsevier.com/locate/jautimm 0896-8411 Ó 2012 Elsevier Ltd. doi:10.1016/j.jaut.2012.04.002 Journal of Autoimmunity 39 (2012) 304e314 Open access under CC BY-NC-ND license. Open access under CC BY-NC-ND license.
  • 2. human EBA [23]. In these experimental models, binding of anti- bodies to its target antigen alone was not sufficient for lesion formation but complement activation, the recruitment and acti- vation of leukocytes, and the release of proteases, that finally led to the dermal-epidermal separation, were essential [24e26]. The central role of the Fc portion of anti-type VII collagen IgG was shown by the lack of pathogenicity of both rabbit F(ab’)2 fragments and chicken IgY against anti-type VII collagen [11,27]. The glycosylation status of the Fc portion of IgG is important for interaction with Fcg receptors (FcgRs). In fact, hydrolysis of IgG glycan reduces the binding ability to FcgRs in both humans and mice [28,29]. EndoS, derived from Streptococcus pyogenes, is an endoglycosidase that hydrolyzes the conserved N-linked glycan on IgG heavy chains [30e32]. To date, EndoS is the only known endoglycosidase that exclusively hydrolyzes the glycan of native IgG [32,33]. Treatment of most autoantibody-mediated diseases, including EBA, is largely unspecific and relies on the long-term use of systemic corticosteroids and additional immunosuppressants that is frequently associated with severe adverse reactions. Thus, novel therapeutic avenues are warranted and indeed, EndoS has previ- ously been successfully applied in experimental autoimmune diseases, including rheumatoid arthritis [34] and immune- mediated thrombocytopenia [32]. However, the effect of EndoS on autoantibodies that have already bound to their target antigens and that have already induced clinical disease has not yet been addressed. Here, we showed that EndoS significantly reduced the patho- genic effect of autoantibodies to type VII collagen in 3 different experimental models. Importantly, closely mimicking the patient situation, EndoS suppressed disease activity in the immunization- induced mouse model, even when applied after anti- collagen type VII antibodies had bound to the skin and clinical disease had developed. Extending the present knowledge of the action of EndoS the enzyme was shown to hydrolyze already in vivo-bound path- ogenic autoantibodies and to differentially modulate the expres- sion of activating and inhibiting FcgRs. 2. Materials and methods 2.1. Generation of rabbit antibodies to murine collagen type VII (mCOL7) Pathogenic IgG was generated as previously described [11]. In brief, New Zealand white rabbits were immunized with recombi- nant forms of the glutathione S-transferase (GST)-tagged NC1 domain of mCOL7 and the rabbit serum was purified by affinity chromatography using protein G. Reactivity of IgG fractions was analyzed by indirect immunofluorescence (IF) microscopy on murine (BALB/c) skin. In parallel, rabbit IgG against GST alone was prepared. 2.2. EndoS preparation and IgG hydrolysis in vitro Recombinant GST-tagged EndoS was expressed in Escherichia coli as previously described [33]. One mg of rabbit anti-mCOL7 IgG was incubated with 5 mg recombinant GST-EndoS in PBS for 16 h at 37 C followed by affinity removal of GST-EndoS by serial passages over Glutathione-Sepharose 4B columns (GE Healthcare, Uppsala, Sweden). This GST-EndoS-treated IgG is called EndoS-pretreated IgG throughout the paper. Prior treatment of animals, the GST-tag was removed from GST-EndoS using factor Xa (Novagen, Madi- son, WI). Liberated GST and factor Xa was removed by serial passage over Xarrest agarose (Novagen) and Glutathione-Sepharose 4B. 2.3. IgG purification, SDS-PAGE, and lectin blotting For sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) and lectin blot analyses, total IgG was purified from 10 ml mouse serum samples using Ab SpinTrap (GE Healthcare) according to manufacturer’s instructions. SDS-PAGE and immuno- blotting were performed as previously described [32]. Glycosylated IgG was detected by using 5 mg/ml biotinylated Lens culinaris agglutinin (LCA)-lectin (Vector Laboratories, Burlingame, CA) and 1 mg/ml streptavidinehorseradish peroxidase (Vector Laboratories) and Super Signal West Pico peroxidase substrate (Pierce, Rockford, IL). Membranes were analyzed by using a Chemidoc XRS imaging system and Quantity One image analysis software (BioRad, Hercules, CA). 2.4. Cryosection assay Skin dermal-epidermal separation was evaluated using an ex vivo model as previously described [35]. Briefly, leukocytes from the peripheral blood of healthy volunteers were isolated by a sedi- mentation gradient containing sodium diatrizoate and dextran 500 following the instructions of the manufacturer (Nycomed, Oslo, Norway), then harvested and washed twice in RPMI 1640 (Life Technologies, Karlsruhe, Germany) and resuspended in the same medium. The cell suspension was kept on ice and cell viability was tested using trypan blue; preparations with viability greater than 95% were used. Cryosections prepared from adult mouse tail skin (BALB/c) were placed in the center of a Superfrost Plus microscope slide (Menzel-Glaser, Braunschweig, Germany). Skin sections were washed with PBS for 5 min to remove embedding medium, then incubated with 50 ml of 1:2 diluted either EndoS-pretreated anti- mCOL7 IgG or non-treated anti-mCOL7 IgG for 90 min at room temperature. After washing the sections with PBS twice, chambers were prepared as described and the leukocyte suspension at the concentration of 1 107 cell/ml was injected into the chambers. Subsequently, chambers were disassembled, and sections were washed in PBS, fixed in formalin, and stained with hematoxylin and eosin. Finally, skin dermal-epidermal separation was evaluated microscopically and the extent expressed as the proportion of the epidermis detached from the dermis. Approval for these studies was obtained from the Institutional Review board at the University of Lübeck (Lübeck, Germany 09-140), and informed consent was provided according to the Declaration of Helsinki. 2.5. Induction of autoimmunity to mCOL7 2.5.1. Animals Six to 8 week-old BALB/c mice for passive EBA model and SJL for active EBA model were purchased from Charles River (Sulzfeld, Germany). Animals were housed at 12 h lightedark cycle at the experimental animal facility in the University of Lübeck. All clinical examinations, biopsies and bleedings were performed under anesthesia using intraperitoneal administration of a mixture of ketamine (100 mg/g) and xylazine (15 mg/g). Animal experiments were approved by local authorities of the Animal Care and Use Committee (Kiel, Germany; 68-6/09) and performed by certified personnel. 2.5.2. Passive transfer model Passive transfer studies followed a previously published protocol [11] with minor modifications. Briefly, mice received 7.5 mg of rabbit anti-mCOL7 IgG or EndoS-pretreated rabbit anti- mCOL7 IgG subcutaneously in abdominal skin every other day from day 0 in total of 6 injections. Alternatively, EndoS was injected i.p. 24 h after anti-mCOL7 IgG injections. Mice were evaluated at 4 M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 305
  • 3. day-intervals for presence of skin lesions until 12 days after first IgG injection (day 12). Cutaneous lesions were counted, and the extent of skin disease was scored as involvement of the skin surface as reported previously [11]. At the same time intervals, blood was obtained for analysis of circulating antibodies. At day 12, all mice were sacrificed, blood was taken, and both lesional and perilesional biopsies were taken for histopathological analysis (stored in 4% buffered formalin) and perilesional biopsies were direct IF microscopy for determination of IgG and C3 deposits at the DEJ (stored at 80 C), respectively. 2.5.3. Immunization-induced model Immunization-induced autoimmunity to mCOL7 was generated as previously reported [36,37]. In brief, SJL mice were immunized at the hind footpad with 60 mg of recombinant murine type VII collagen immunodominant peptide emulsified in the non-ionic block copolymer adjuvant TiterMax (ALEXIS Biochemicals, Lör- rach, Germany). When mice had developed disease with a clinical score greater 2, they were randomly allocated into 3 experimental groups (week 0). Two groups were injected intraperitoneally with 200 mg EndoS and 400 mg EndoS, respectively, once a week for 4 weeks. Mice were clinically evaluated at a week intervals for presence of skin lesions until week 4; at the same time blood was taken. Cutaneous lesions were counted, and the extent of skin disease was scored as involvement of the skin surface as reported previously [23]. At the end of week 4, biopsies were taken and blood was drawn as described above for the passive transfer model. Circulating autoantibodies against mCOL7 were detected by ELISA as previously described [37]. 2.6. Histopathology Formalin-fixed skin samples were processed into paraffin blocks. 4 mm sections were stained with hematoxylin and eosin according to standard protocols [37]. 2.7. Immunohistochemistry Six mm cryosections of lesional skin obtained from mice were fixed in acetone at 20 C for 10 min. Air dried sections were washed with PBS for 5 min 3 times and incubated with Dako Dual Endogenous Enzyme Block (DakoCytomation, Hamburg, Germany) Fig. 1. EndoS-treated pathogenic IgG did not induce separation at the dermal-epidermal junction (DEJ) of normal mouse skin ex vivo. Six mm cryosections incubated with non- treated (a) and EndoS-pretreated (b) rabbit anti-mCOL7 IgG. After addition of granulocytes from healthy volunteers for a 1 h incubation time, significantly more neutrophils were recruited along the DEJ in sections incubated with untreated compared to sections incubated with EndoS-treated rabbit anti-mCOL7 IgG (c; ***p 0.001, t-test). White triangles (D) indicate the DEJ. After an incubation period of 3 h, significantly less subepidermal split formation was seen in sections incubated with untreated rabbit anti-mCOL7 IgG (d) compared to sections incubated with EndoS-treated rabbit anti-mCOL7 IgG (e). The size of separation at the DEJ is significantly less in EndoS-pretreated rabbit anti-mCOL7 IgG incubated sections compared with non-treated rabbit anti-mCOL7 IgG (f). *p 0.05 (t-test). The experiment was conducted six times in triplicate for the evaluation of the DEJ separation. For the neutrophil binding evaluation, one experiment in triplicate was performed. The extent of neutrophil binding at the DEJ of the two random areas of the entire section with high power view (400) and of DEJ separation was evaluated by two blinded observers. M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 306
  • 4. for 30 min at room temperature (RT) for blocking endogenous peroxidase. To reduce non-specific binding of secondary antibody, sections were incubated with 5% normal rabbit serum for 20 min at RT. After excess serum from sections was blotted, sections were incubated with rat anti-mouse Ly-6C/ly-6G (Gr-1; RB56-8C5, BD Pharmingen, San Diego, CA, USA) antibody (1:200 dilution in 2% normal rabbit serum/PBS) for 1 h at RT. Slides were washed with PBS followed by incubation with rabbit anti-rat IgG (1:200 dilution in 2% normal rabbit serum/PBS; DakoCytomation) for 45 min at RT. After washing, sections were incubated with R.T.U. VECTASTAINÒ Elite ABC Reagent (Vector Laboratories) for 30 min at RT. To develop the peroxidase reaction, sections were incubated in HistoGreen (LINARIS, Wertheim-Bettingen, Germany) solution until appro- priate color developed. Excess solution was washed in tap water, followed by counterstaining with hematoxylin and sections were mounted. 2.8. Immunofluorescence (IF) microscopy Direct IF microscopy to detect tissue-bound IgG, complement C3 deposits has been described previously [23]. Staining intensity of immunoreactants at the DEJ was quantified with ImageJ software (http://rsbweb.nih.gov/ij/). Six mm sections of healthy BALB/c mouse skin were prepared for indirect IF microscopy to evaluate tissue binding ability of rabbit anti-mCOL7 IgG and mouse sera [37]. Binding of autoantibodies and C5 to the skin was visualized using FITC-conjugated rabbit anti-mouse IgG and the monoclonal anti- mouse C5 antibody BB5.1, respectively (Dako and Cell Sciences, Canton, MA, USA). 2.9. Quantitive reverse transcriptase-polymerase chain reaction (RT-PCR) To evaluate the FcgR and inflammatory cytokine expression in the skin, total RNA was isolated from the half of the right ear according to the manufacturer’s protocol (Analytic Jena Ag, Neu Ruppin, Germany). The RNA was incubated with DNase I (SigmaeAldrich Chemie, Munich, Germany) for 15 min and fol- lowed by heating at 70 C for 10 min, then immediately cooled on ice. After reverse transcription the cDNA was added to the qPCR Master Mix Plus SYBR Green I (Eurogentec, Cologne, Germany) and amplified using the SDS ABI 7900 system (Applied Biosystems, Darmstadt, Germany). The forward and reverse primes were designed by using the computer software CloneManager (Sci Ed Central, Version7.01; Cary, NC, USA). The optimal primmer concentrations used were 500 nM each for the forward and reverse primers (biomers.net, Ulm, Germany). Primers used for this project are shown in Supplementary Table 1. The same batch of cDNA (20 ml) was used to determine the cycles of threshold (ct), and the amount of the cytokine cDNA copies were normalized to the house keeping gene MLN51. 2.10. Autoantibody elution from the skin Mice (n ¼ 4) were injected twice with rabbit anti-mCOL7 IgG at day 0 and 2. On day 4, two mice were injected with EndoS 400 mg and the other two mice remained untreated. Twenty-four hours later (day 5) mice were sacrificed and skin was sampled and at 80 C. Ten mm sections were prepared and placed on Superfrost Plus microscope slides (Menzel-Glaeser). After removing bedding medium by washing with PBS, antibodies were eluted from the skin washing with 0.1M glycin, pH 2.8 at 4 C as described previously [38]. The collected antibodies were neutralized with TriseHCl and concentrated (Millipore, Cork, Ireland and Vivaspin 500, Sartorius Stedim, Goettingen, Germany). Autoantibody binding was verified by indirect IF microscopy on mouse skin. 2.11. Statistical analysis Sigma plot 11.0 (Systat Software, Inc, Chicago, USA) was used to perform statistical analysis. A p-value of 0.05 was considered statistically significant. Applied tests are indicated at the respective table or figure legends. cl l ∗∗ ∗∗∗ ∗∗∗ b c a r Fig. 2. EndoS-pretreatment of anti-mCOL7 IgG failed to induce clinical disease in vivo. BALB/c mice were subcutaneously injected with 7.5 mg per head EndoS-treated (n ¼ 8) and untreated rabbit anti-mCOL7 IgG (n ¼ 11) every other day over 12 days (total 45 mg). Clinical scores were evaluated every 4 days and determined as the extent of body surface affected by lesions. Mice injected with EndoS-pretreated rabbit anti-mCOL7 IgG demonstrate a significantly lower clinical score compared with mice that received untreated rabbit anti-mCOL7 IgG alone (a; ***p 0.001, **p 0.01, t-test). Representative clinical pictures of mice injected with untreated rabbit mCOL7 IgG (b) or EndoS-pretreated rabbit anti- mCOL7 IgG (c) at day 12. M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 307
  • 5. 3. Results 3.1. Hydrolyzed anti-mouse type VII collagen IgG did not change the binding capacity to its target antigen in vitro The IgG glycan hydrolyzing effect of EndoS treatment was verified by SDS-PAGE and Lens culinaris agglutinin (LCA) lectin blot analysis before EndoS-treated IgG was applied in further experi- ments. After successful IgG glycan hydrolysis, SDS-PAGE and LCA lectin blot analysis showed a reduction of the molecular size of the heavy chain of rabbit anti-mouse type VII collagen (mCOL7) IgG by approximately 3 kDa and a strongly reduced signal of the EndoS- treated rabbit anti-mCOL7 IgG compared to untreated IgG, respectively (Supplementary Fig. 1). Serial dilutions of EndoS- treated and untreated anti-mCOL7 IgG used in equal concentra- tions resulted in the same titer (1:51,200) and binding pattern by indirect immunofluorescence (IF) microscopy on mouse skin (data not shown). Thus, hydrolyzation by EndoS did not affect the F(ab’)2 fragment function, i.e. the binding of anti-anti-mCOL7 IgG to the dermal-epidermal junction (DEJ). 3.2. EndoS-pretreatment of otherwise pathogenic IgG failed to induce dermal-epidermal separation ex vivo To evaluate the effect of EndoS treatment, cryosections of normal mouse skin were incubated in vitro with EndoS-treated and untreated rabbit anti-mCOL7 IgG, respectively. After addition of granulocytes from healthy volunteers for a 1 h incubation time, significantly more neutrophils were recruited along the DEJ in sections incubated with untreated compared to sections incubated with EndoS-treated anti-mCOL7 IgG (Fig. 1aec; p 0.001). After an incubation period of 3 h, significantly less subepidermal split formation was observed in sections incubated with EndoS-treated IgG compared to sections incubated with untreated anti-mCOL7 IgG (Fig. 1def; p 0.05). 3.3. EndoS-pretreatment of otherwise pathogenic IgG failed to induce clinical disease in vivo Since the ex vivo pathogenicity of rabbit anti-mCOL7 IgG was abrogated after treatment with EndoS, the next set of experiences addressed the in vivo effect of EndoS treatment. BALB/c mice were subcutaneously injected with 7.5 mg per head EndoS-treated (n ¼ 8) and untreated anti-mCOL7 IgG (n ¼ 11) every other day over 12 days. Mice injected with EndoS-treated IgG (Fig. 2c) did not develop clinical disease while, as expected, mice injected with untreated IgG (Fig. 2b) developed severe skin lesion reproducing the human disease (Fig. 2a). These data were obtained from 2 independent experiments with different anti-mCOL7 IgG preparations. 3.4. Treatment of EndoS following injection of pathogenic IgG suppressed disease severity in vivo To evaluate the effect of EndoS in vivo, BALB/c mice received intraperitoneal EndoS every other day, starting at day 1 until day 11, at a dose of 10 mg/injection (n ¼ 8) or 100 mg/injection (n ¼ 8) 24 h after anti-mCOL7 IgG injection. Disease severity was significantly milder in mice that had received 100 mg EndoS in conjunction with anti-mCOL7 IgG compared to both mice that had been injected with 10 mg EndoS plus anti-mCOL7 IgG and mice that had received anti- mCOL7 IgG alone (Fig. 3a). The IgG glycan status of mouse serum was analyzed in all mice at day 12. SDS-PAGE and LCA lectin blot- ting confirmed that IgG from all EndoS-treated mice was signifi- cantly less glycosylated compared to IgG from mice that had not been treated with EndoS (Fig. 3b). 3.5. Autoantibody-mediated intralesional inflammatory responses were inhibited by EndoS The inflammatory response in skin lesions of mice was subse- quently analyzed by histopathology and quantification of mRNA for different cytokines, chemokines, and cellular markers, including ∗∗ a b cl l r Fig. 3. Treatment of EndoS following injection of pathogenic IgG suppressed disease severity in vivo. EndoS was applied at a dose of 10 mg/injection (n ¼ 8) or 100 mg/ injection (n ¼ 8), respectively, every other day, starting at day 1 until day 11, 24 h after rabbit anti-mCOL7 IgG injection. Disease severity was significantly milder in mice that had received 100 mg EndoS in conjunction with rabbit anti-mCOL7 IgG compared to mice that had received rabbit anti-mCOL7 IgG alone. (a; **p 0.01, One-way ANOVA). The IgG glycan status of serum was analyzed in all mice obtained at day 12. SDS-PAGE (Stain) and Lens culinaris agglutinin (LCA) lectin blotting confirmed that in the 100 mg EndoS group and, to a lesser extent, in the 10 mg Endo group, IgG was considerably less hydrolyzed compared to IgG from untreated mice. Results form 8 individual mice of each group are shown. Blots were performed in the same experiment including the appropriate controls and were rearranged for better comparison (b). M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 308
  • 6. FcgR, by RT-PCR. By histopathology of biopsies from mouse ears, significantly less inflammatory cells were observed in the dermis of both mice that had received EndoS-pretreated anti-mCOL7 IgG (n ¼ 8) and mice had been injected with EndoS along with anti- mCOL7 IgG (n ¼ 8) compared to mice that were injected with anti-mCOL7 IgG alone (n ¼ 11; Fig. 4aec). Also, mRNA levels of T cell (CD3), dendritic cell (CD11c), macrophage and monocyte markers (Mac-1) as well as of chemokines (KC, MIP) and inflammatory cytokines (TNFa, and IL-1b) were significantly lower in skin of mice that had received EndoS-pretreated anti-mCOL7 IgG (n ¼ 8) compared to mice that had obtained untreated anti-mCOL7 IgG (n ¼ 11; p 0.001 for CD3, KC, MIP, and IL-1b, p 0.05 for CD11c, p 0.01 for TNFa; Fig. 4d). In contrast, mRNA levels of IFN-g, RANTES, and IL-6 remained unaltered (p ¼ 0.570, p ¼ 0.483 and p ¼ 0.06, respectively). 3.6. Autoantibody-mediated in vivo deposition of IgG and complement was unaltered by EndoS In order to investigate the effect of EndoS on the binding of autoantibodies to their target antigen and subsequent complement recruitment, in vivo deposition of IgG, C3, and C5 at the DEJ were visualized by direct IF microscopy of mouse ear skin obtained from the three experimental groups: anti-mCOL7 IgG alone (Fig. 5aec), EndoS-pretreated anti-mCOL7 IgG (Fig. 5def), and EndoS in conjunction with anti-mCOL7 IgG (gei). The intensity of fluores- cence was quantified using ImageJ software. The levels of tissue- bound IgG (Fig. 5j), C3 (Fig. 5k), and C5 (Fig. 5l) were not signifi- cantly different between the three experimental group. These data indicate the EndoS did not interfere with autoantibody binding and complement recruitment in vivo. 3.7. Therapeutic efficacy of EndoS in immunization-induced autoimmunity To mimic the therapeutic situation in patients with autoantibody-mediated disorders more closely, mice (n ¼ 50) were immunized with a recombinant portion of the immunodominant NC1 domain of murine type VII collagen. Mice that developed a disease score of greater 2 (between 4 and 8 weeks after immu- nization) were allocated to three groups: no treatment (n ¼ 16), i.p. injection of 200 mg (n ¼ 11) and 400 mg (n ¼ 12) EndoS once per week for 4 weeks, respectively. Mice in both EndoS treatment groups developed significantly less disease compared to non- treated mice (Fig. 6aec). Importantly, disease progression was already abrogated after the first treatment week in the high-dose group pointing to a direct effect of EndoS on the glycosylation status of in vivo-bound autoantibody. In addition, the number of mice in which disease activity decreased below the clinical score at the time of treatment allocation was significantly higher in the EndoS groups (11 of 23) compared to the untreated mice (0 of 16; p ¼ 0.00065, Fisher’s exact test). Histopathological analysis showed significantly less infiltration of inflammatory cells including Gr-1 positive cells in dermal lesions of EndoS-treated mice compared Fig. 4. Autoantibody-mediated intralesional inflammatory responses were inhibited by EndoS. Evaluation of the inflammatory response in ear skin was performed by histopa- thology (aec) and quantification of mRNA for different chemokines, cytokines, and cell markers by RT-PCR (d). Representative histopathology pictures of skin obtained from mice injected with untreated anti-mCOL7 IgG alone demonstrates significantly higher inflammatory cell infiltration (a) compared with skin from mice injected with EndoS-pretreated anti-mCOL7 IgG (b) or untreated anti-mCOL7 IgG together with 100 mg of EndoS (c). By RT-PCR, mRNA levels of makers for T cells (CD3), dendritic cell (CD11c) and macrophages/ monocytes (Mac-1) as well as of chemokines (KC and MIP) and inflammatory cytokines (TNFa and IL-1b) were significantly less in skin of mice that received EndoS-pretreated anti- mCOL7 IgG (n ¼ 8) compared to that of untreated anti-mCOL7 IgG-injected mice (n ¼ 11; d; *p 0.05, **p 0.01, ***p 0.001, t-test). M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 309
  • 7. to untreated animals (Fig. 6fei). By direct IF microscopy, IgG and C3 deposits at the DEJ did not differ between any of the groups (Fig. 6jem). 3.8. EndoS does hydrolyze already-tissue-bound autoantibodies Since EndoS injection reversed already established clinical disease we further evaluated if EndoS could hydrolyze autoanti- bodies that had already bound to their target antigen in the tissue. IgG that was eluted from the skin following binding of pathogenic anti-mCOL7 IgG and subsequent EndoS treatment showed size reduction by SDS-PAGE and was unreactive by lectin blotting in contrast to IgG eluted from the skin of mice that did not receive EndoS (Fig. 7). Both eluted IgG fractions equally labeled the DEJ of mouse skin by indirect IF microscopy (Fig. 7). 3.9. EndoS modulates the expression of Fcg receptors in autoantibody-mediated skin destruction In mice that received EndoS-pretreated anti-mCOL7 IgG, mRNA levels of the activating FcgRs were significantly reduced compared to mice injected with untreated anti-mCOL7 IgG (Fig. 8a). In Fig. 5. Autoantibody-mediated in vivo deposition of IgG and complement was unaltered by EndoS. By direct immunofluorescence microscopy of non-lesional mouse skin obtained from mice injected with anti-COL7 IgG (a, b, c), EndoS-pretreated anti-COL7 IgG (d, e, f), and anti-mCOL7 IgG together with 100 mg of EndoS (g, h, i), in vivo deposits of IgG (a, d, g), C3 (b, e, h) and C5 (c, f, i) at the dermal-epidermal junction were visualized. The intensity of fluorescence was quantified using ImageJ software (j, IgG; k, C3; l, C5). No significant difference was observed between the 3 groups. M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 310
  • 8. contrast, no differences were seen in mRNA levels of the inhibitory FcgRIIB between the two groups (Fig. 8a). Similar data were ob- tained when FcgR expression was related to the CD11c expression in lesional skin, however, statistical significance was not reached (Fig. 8b). When FcgR expression was analyzed in the immunization- induced model, EndoS injections resulted in reduced expression of the activating FcgR and increased expression of FcgRIIB (Fig. 8c). When related to the lesional expression of CD11c, a statistical significant downregulation was observed for FcgRIII and FcgRIV whereas expression of FcgRIIB was significantly increased (Fig. 8d). 4. Discussion EndoS has been identified as glycosidase that specifically hydrolyzes the conserved N-linked glycan on IgG heavy chains [30e32]. EndoS has a unique specificity for native IgG or IgG Fc portion glycans compared to related endoglycosidases which are specific for certain glycans that can be attached to any protein or peptide, and which are enhanced by or require denaturation of the protein backbone [32,33]. By hydrolysis of IgG glycan, binding affinity of the Fc portion of IgG to Fcg receptors (FcgRs) is modu- lated. More precisely, EndoS-treated IgG has a considerably lower affinity to FcgRI, FcgRIIB, FcgRIII, and FcgRIV in mice [29]. Inter- estingly, EndoS-hydrolyzed human IgG2 binds better to human inhibitory receptor FcRIIB, while binding of all EndoS-hydrolyzed IgG subclasses to activating receptors is lowered [28]. The thera- peutic efficacy of EndoS has previously been demonstrated in different experimental autoimmune diseases induced by passive transfer of antibodies, including rheumatoid arthritis [29,34], immune-mediated thrombocytopenia [32], hemolytic anemia [39], and ANCA-mediated glomerulonephritis [40]. Since EndoS does not specifically target pathogenic autoantibodies but hydrolyzes the entire pool of IgG, it may increase the risk for infections and interfere with vaccination-induce immunity in human patients. On the other hand, EndoS does not affect IgG’s F(ab)’ binding capacity and activation of the classical complement system [32] leaving intact large parts of the humoral immune defense system. Furthermore, repetitive injections with EndoS can raise EndoS- specific antibodies which may hamper its efficacy. Here, we extended the current knowledge about the mecha- nisms of action of EndoS by the use of an immunization-induced mouse model in which EndoS exerted its effect on autoantibodies that had already bound to their autoantigen. In addition, expression of the activating FcgRIII and FcgRIV in the skin lesions was down- regulated whereas expression of the inhibiting FcgRIIB was upre- gulated following EndoS treatment. In the first set of experiments, pretreatment of pathogenic anti-mCOL7 IgG with EndoS resulted in a statistically significant reduction of dermal-epidermal separation in cryosections of mouse Fig. 6. Therapeutic efficacy of EndoS in immunization-induced autoimmunity. Mice that developed a disease score of greater 2 (between 4 and 8 weeks after immunization with a recombinant portion of the immunodominant NC1 domain of murine type VII collagen) were allocated into three groups: no treatment (n ¼ 16) and i.p. injection of 200 mg (n ¼ 11) or 400 mg (n ¼ 12) EndoS once per week for 4 weeks, respectively. Mice in both EndoS treatment groups developed significantly less disease compared to non-treated mice (a; *p 0.05). Representative clinical (bee), histo- pathological (f,g,h,i), and immunopathological pictures (jem) of untreated mice (b, f, h, j, l) and 400 mg EndoS-treated mice (c, e, g, i, k, m) are shown. Ear of a mouse that did not receive EndoS (b) demonstrates more clinical disease, e.g. erythema and erosion, compared with that was treated with 100 mg EndoS for 4 weeks (c). Clinical lesion on a mouse tail at week 0 (before treatment, d) was completely healed after 4 weeks of 400 mg EndoS treatment (e). Histopathology of 400 mg EndoS-treated mice (g) shows significantly less infiltration of inflammatory cells including Gr-1 positive cells (gran- ulocytes and monocytes) demonstrated by immunohistochemistry (i) in dermal lesions compared with those of untreated mice (f, h). By direct IF microscopy, IgG (j, k) and C3 (l, m) deposits at the DEJ did not differ between non-treated (j, l) and EndoS- treated (k, m) groups. M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 311
  • 9. skin. In this ex vivo model, cryosections of mouse skin were incu- bated with anti-mCOL7 IgG and subsequently with leukocytes from healthy volunteers. Leukocytes attached to the DEJ and via the release of elastase and matrix metalloproteinase-9 from gran- ulocytes, the DEJ is split [26]. Based on these findings, in the next set of experiments, a mouse model was used in which the injection of anti-mCOL7 IgG reproduces the clinical and immunopathological characteristics of the human disease [11]. In this model, the pretreatment of pathogenic anti-mCOL7 IgG with EndoS prevented clinical disease in all mice. When EndoS was administrated to mice 24 h after the injection of pathogenic anti-mCOL7 IgG, disease activity was significantly reduced. In order to control for a potential effect of EndoS on the interaction of anti-mCOL7 IgG with mCOL7, Fig. 7. EndoS hydrolyzes tissue-bound autoantibodies IgG eluted from the skin of mice that had received anti-mCOL7 IgG followed by injection of EndoS (þ) showed size reduction by Coomassie Brilliant Blue staining (Stain) and lost reactivity by blotting with LCA lectin (LCA blot). In comparison, IgG eluted from the skin of mice that were injected with anti-mCOL7 IgG alone is shown (). Both eluted IgG fraction labeled the dermal-epidermal junction by indirect immunofluorescence microscopy on normal murine skin (IF microscopy). a b c d Fig. 8. EndoS differently modulates the expression of activating and inhibitory Fcg receptors mRNA expression of Fc gamma receptors (FcgRs) in the lesional skin was determined. Expression of activating FcgRs (FcgRI, FcgRIII, and FcgRIV) in EndoS-pretreated anti-mCOL7 IgG-injected mice was significantly less compared with untreated pathogenic IgG- injected animals (a; *p 0.05, t-test). Expression of FcgRIIB was significantly increased following EndoS treatment (a; *p 0.05, t-test). Similar data were obtained in the immunization-induced mouse model (c). Here, data reached statistical significance only for FcgRIV (c; *p 0.05, t-test). When the lesional FcgR expression was related to the expression of CD11c in both experimental models, a decrease of the activating and increase of the inhibitory FcgR(s) was seen (b, d). Statistical significance was observed in the immunization-induced model (d; *p 0.05, t-test). M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 312
  • 10. the binding of anti-mCOL7 IgG to mouse skin was visualized by direct IF microscopy. No significant difference between skin of mice that received anti-mCOL7 IgG alone, EndoS-treated anti-mCOL7 IgG, and pathogenic IgG, followed by EndoS injections, was observed. Furthermore, the binding of C3 and C5 to the DEJ remained unaltered between the 3 groups. Previously, the binding of C3 and C5 to the DEJ and the activation of the alternative pathway were shown to be critical for disease development in this model [24]. EndoS has previously been shown to prevent binding of C1q, a component of the classical pathway, to IgG-sensitized red blood cells and subsequent hemolysis [39]. These data suggest that EndoS may interfere with the binding of IgG to components of the clas- sical, but not of the alternative pathway of complement activation. In autoimmunity to mCOL7, complement deposition at the DEJ is thought to be an essential step to attract inflammatory cells that then become activated via the interaction between the Fc portion of autoantibody and FcgR, which further enhances the local inflam- mation, and finally leads to the release of proteases at the DEJ. Regarding the pathogenic role of complement, we observed unchanged levels of tissue-bound C3 and C5. It has been proposed that anaphylatoxin C5a, which does not bind to tissue, and C5aR on neutrophils may compose an amplification loop for neutrophil activation in ANCA-mediated nephritis model [41]. This speculates that the less neutrophils due to inhibited the binding of FcgRs on neutrophils and Fc part of the pathogenic IgG caused less C5a production, and eventually reduce the disease severity at the lesion. Our results indicate that EndoS treatment interferes with this further enhancement of the local inflammation, thus inhibiting dermal-epidermal separation. To mimic the situation of EBA patients more closely, in the last set of experiments, an immunization-induced model was employed. In this model, SJL mice develop the clinical and immunopathological hallmarks of the human disease 4e8 weeks after immunizationwith a recombinant fragment of the immunodominant NC1 domain of mCOL7 [23]. This mouse model appeared to be particularly attrac- tive since development of clinical disease depended on the depo- sition of IgG2b autoantibodies to the DEJ [23] and EndoS was shown to only functionally impair IgG1 and IgG2b subclasses in mice [34]. In the present study, after disease onset, mice were allocated to 3 groups: 2 groups received weekly injections of different EndoS doses and the third group was left untreated. In both treatment groups, progression of the disease was abrogated. In fact, clinical disease was reduced in nearly half of EndoS-treated mice but in none of untreated animals. Importantly, in the high-dose EndoS group, progression of the clinical disease activity was already inhibited one week after the first EndoS injection. In the low-dose EndoS group, this effect was observed one week later. Since anti-mCOL7 IgG bound in murine skin was previously found to remain in place for about 8 weeks by investigating the off-spring of mice immunized with recombinant mCOL7 [36], the clinical effect of EndoS in the present study is most likely explained by a functional interaction of EndoS with tissue- bound anti-mCOL7 IgG. This assumption was experimentally corroborated when skin-bound anti-mCOL7 IgG eluted after in vivo EndoS treatment was shown to be hydrolyzed in contrast to eluted anti-mCOL7 IgG without preceding EndoS treatment. These find- ings have implications on a future therapeutic use of EndoS in autoantibody-mediated diseases in which EndoS may interfere not only with serum autoantibodies but also with autoantibodies that have already bound to their target tissue. Finally, we explored the effect of EndoS on the composition of the inflammatory infiltrate and the differential expression of FcgR in skin. We found mRNA levels of the different FcgR in the mouse skin not to be equally affected by EndoS treatment, which has not previously been reported. In both the passive transfer model and immunization-induced model, the activating FcgRs, including FcgRI and low affinity FcgRIII and FcgRIV were significantly downregulated, while the inhibiting FcgRIIB was upregulated following EndoS treatment. This effect was shown to be in part attributed to the reduction of the inflammatory infiltrate in the skin lesions. In addition, a novel independent effect of EndoS on the FcgR expression was observed. The pathogenic relevance of upre- gulated activating FcgRs and downregulated FcgRIIB has previously been shown in autoimmunity including human and experimental rheumatoid arthritis, experimental arthritis and immune-complex mediated nephritis [42,43] as well as, most recently, autoimmunity to type VII collagen [44]. In conclusion, we extended the present knowledge about the mechanisms of action of EndoS and favor the further exploration of enzymatic autoantibody glycan hydrolysis as novel, therapeutic option in severe autoantibody-mediated disease. Conflicts of interest disclosure Patents for the in vitro and in vivo use of EndoS have been applied for by Genovis AB and Hansa Medical AB, respectively. M.C. is listed as an inventor on these applications that are pending. Hansa Medical AB in part funded this study, but had no influence on the design of study, interpretation of data, or the final form of the manuscript. M.C. is a part time scientific consultant for Hansa Medical AB. Acknowledgments Ulla Johannesson, Lund, and Rebecca Cames and Claudia Kau- derer, Lübeck, are acknowledged for excellent technical assistance. This work was supported by grants from the Excellence Cluster “Inflammation at Interfaces” (EXC306/1), Novartis Pharma (to E.S.) as well as the Swedish Research Council (projects 2005-4791 and 2010-57X-20240), the Foundations of Crafoord, Hedberg, Bergvall, Österlund, Wiberg, Söderberg, the Swedish Society for Medicine, the Royal Physiografic Society, King Gustaf V’s Memorial Fund, the Swedish Rheumatism Association, the Medical Faculty at Lund University, and Hansa Medical AB to M.C. Appendix A. Supplementary material Supplementary data related to this article can be found, in the online version at, doi:10.1016/j.jaut.2012.04.002. References [1] Lim PL, Zouali M. Pathogenic autoantibodies: emerging insights into tissue injury. Immunol Lett 2006;103:17e26. [2] Nandakumar KS, Holmdahl R. Therapeutic cleavage of IgG: new avenues for treating inflammation. Trends Immunol 2008;29:173e8. [3] Stanley JR, Amagai M. Pemphigus, bullous impetigo, and the staphylococcal scalded-skin syndrome. N Engl J Med 2006;355:1800e10. [4] Schmidt E, Zillikens D. Modern diagnosis of autoimmune blistering skin diseases. Autoimmun Rev 2010;10:84e9. [5] Sticherling M, Erfurt-Berge C. Autoimmune blistering diseases of the skin. Autoimmun Rev; 2011. [6] Woodley DT, Briggaman RA, O’Keefe EJ, Inman AO, Queen LL, Gammon WR. Identification of the skin basement-membrane autoantigen in epidermolysis bullosa acquisita. N Engl J Med 1984;310:1007e13. [7] Woodley DT, Burgeson RE, Lunstrum G, Bruckner-Tuderman L, Reese MJ, Briggaman RA. Epidermolysis bullosa acquisita antigen is the globular carboxyl terminus of type VII procollagen. J Clin Invest 1988;81:683e7. [8] Labib RS, Anhalt GJ, Patel HP, Mutasim DF, Diaz LA. Molecular heterogeneity of the bullous pemphigoid antigens as detected by immunoblotting. J Immunol 1986;136:1231e5. [9] Giudice GJ, Emery DJ, Diaz LA. Cloning and primary structural analysis of the bullous pemphigoid autoantigen BP180. J Invest Dermatol 1992;99:243e50. M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 313
  • 11. [10] Chen M, Keene DR, Costa FK, Tahk SH, Woodley DT. The carboxyl terminus of type VII collagen mediates antiparallel dimer formation and constitutes a new antigenic epitope for epidermolysis bullosa acquisita autoantibodies. J Biol Chem 2001;276:21649e55. [11] Sitaru C, Mihai S, Otto C, Chiriac MT, Hausser I, Dotterweich B, et al. Induction of dermal-epidermal separation in mice by passive transfer of antibodies specific to type VII collagen. J Clin Invest 2005;115:870e8. [12] Liu Z, Diaz LA, Troy JL, Taylor AF, Emery DJ, Fairley JA, et al. A passive transfer model of the organ-specific autoimmune disease, bullous pemphigoid, using antibodies generated against the hemidesmosomal antigen, BP180. J Clin Invest 1993;92:2480e8. [13] Nishie W, Sawamura D, Goto M, Ito K, Shibaki A, McMillan JR, et al. Human- ization of autoantigen. Nat Med 2007;13:378e83. [14] Recke A, Sitaru C, Vidarsson G, Evensen M, Chiriac MT, Ludwig RJ, et al. Pathogenicity of IgG subclass autoantibodies to type VII collagen: induction of dermal-epidermal separation. J Autoimmun 2010;34:435e44. [15] Sakai LY, Keene DR, Morris NP, Burgeson RE. Type VII collagen is a major structural component of anchoring fibrils. J Cell Biol 1986;103:1577e86. [16] Wetzels RH, Robben HC, Leigh IM, Schaafsma HE, Vooijs GP, Ramaekers FC. Distribution patterns of type VII collagen in normal and malignant human tissues. Am J Pathol 1991;139:451e9. [17] Lapiere JC, Woodley DT, Parente MG, Iwasaki T, Wynn KC, Christiano AM, et al. Epitope mapping of type VII collagen. Identification of discrete peptide sequences recognized by sera from patients with acquired epidermolysis bullosa. J Clin Invest 1993;92:1831e9. [18] Gammon WR, Murrell DF, Jenison MW, Padilla KM, Prisayanh PS, Jones DA, et al. Autoantibodies to type VII collagen recognize epitopes in a fibronectin- like region of the noncollagenous (NC1) domain. J Invest Dermatol 1993;100: 618e22. [19] Tanaka T, Furukawa F, Imamura S. Epitope mapping for epidermolysis bullosa acquisita autoantibody by molecularly cloned cDNA for type VII collagen. J Invest Dermatol 1994;102:706e9. [20] Sitaru C, Kromminga A, Hashimoto T, Brocker EB, Zillikens D. Autoantibodies to type VII collagen mediate Fcgamma-dependent neutrophil activation and induce dermal-epidermal separation in cryosections of human skin. Am J Pathol 2002;161:301e11. [21] Woodley DT, Chang C, Saadat P, Ram R, Liu Z, Chen M. Evidence that anti-type VII collagen antibodies are pathogenic and responsible for the clinical, histological, and immunological features of epidermolysis bullosa acquisita. J Invest Dermatol 2005;124:958e64. [22] Woodley DT, Ram R, Doostan A, Bandyopadhyay P, Huang Y, Remington J, et al. Induction of epidermolysis bullosa acquisita in mice by passive transfer of autoantibodies from patients. J Invest Dermatol 2006;126:1323e30. [23] Sitaru C, Chiriac MT, Mihai S, Buning J, Gebert A, Ishiko A, et al. Induction of complement-fixing autoantibodies against type VII collagen results in subepidermal blistering in mice. J Immunol 2006;177:3461e8. [24] Mihai S, Chiriac MT, Takahashi K, Thurman JM, Holers VM, Zillikens D, et al. The alternative pathway of complement activation is critical for blister induction in experimental epidermolysis bullosa acquisita. J Immunol 2007; 178:6514e21. [25] Chiriac MT, Roesler J, Sindrilaru A, Scharffetter-Kochanek K, Zillikens D, Sitaru C. NADPH oxidase is required for neutrophil-dependent autoantibody- induced tissue damage. J Pathol 2007;212:56e65. [26] Shimanovich I, Mihai S, Oostingh GJ, Ilenchuk TT, Brocker EB, Opdenakker G, et al. Granulocyte-derived elastase and gelatinase B are required for dermal- epidermal separation induced by autoantibodies from patients with epi- dermolysis bullosa acquisita and bullous pemphigoid. J Pathol 2004;204: 519e27. [27] Sesarman A, Mihai S, Chiriac MT, Olaru F, Sitaru AG, Thurman JM, et al. Binding of avian IgY to type VII collagen does not activate complement and leucocytes and fails to induce subepidermal blistering in mice. Br J Dermatol 2008;158: 463e71. [28] Allhorn M, Olin AI, Nimmerjahn F, Collin M. Human IgG/Fc gamma R inter- actions are modulated by streptococcal IgG glycan hydrolysis. PLoS One 2008; 3:e1413. [29] Nandakumar KS, Collin M, Olsen A, Nimmerjahn F, Blom AM, Ravetch JV, et al. Endoglycosidase treatment abrogates IgG arthritogenicity: importance of IgG glycosylation in arthritis. Eur J Immunol 2007;37:2973e82. [30] Collin M, Olsen A. EndoS, a novel secreted protein from Streptococcus pyo- genes with endoglycosidase activity on human IgG. EMBO J 2001;20: 3046e55. [31] Collin M, Svensson MD, Sjoholm AG, Jensenius JC, Sjobring U, Olsen A. EndoS and SpeB from Streptococcus pyogenes inhibit immunoglobulin-mediated opsonophagocytosis. Infect Immun 2002;70:6646e51. [32] Collin M, Shannon O, Bjorck L. IgG glycan hydrolysis by a bacterial enzyme as a therapy against autoimmune conditions. Proc Natl Acad Sci U S A 2008;105: 4265e70. [33] Collin M, Olsen A. Effect of SpeB and EndoS from Streptococcus pyogenes on human immunoglobulins. Infect Immun 2001;69:7187e9. [34] Albert H, Collin M, Dudziak D, Ravetch JV, Nimmerjahn F. In vivo enzymatic modulation of IgG glycosylation inhibits autoimmune disease in an IgG subclass-dependent manner. Proc Natl Acad Sci U S A 2008;105:15005e9. [35] Sitaru C, Schmidt E, Petermann S, Munteanu LS, Brocker EB, Zillikens D. Auto- antibodies to bullous pemphigoid antigen 180 induce dermal-epidermal separation in cryosections of human skin. J Invest Dermatol 2002;118:664e71. [36] Kasperkiewicz M, Hirose M, Recke A, Schmidt E, Zillikens D, Ludwig RJ. Clearance rates of circulating and tissue-bound autoantibodies to type VII collagen in experimental epidermolysis bullosa acquisita. Br J Dermatol 2010; 162:1064e70. [37] Ludwig RJ, Recke A, Bieber K, Muller S, Marques Ade C, Banczyk D, et al. Generation of antibodies of distinct subclasses and specificity is linked to H2s in an active mouse model of epidermolysis bullosa acquisita. J Invest Dermatol 2011;131:167e76. [38] Korman NJ. In situ-bound antibodies eluted from the skin of patients with bullous pemphigoid are preferentially directed against the 230-kD bullous pemphigoid antigen. J Invest Dermatol 1995;105:824e30. [39] Allhorn M, Briceno JG, Baudino L, Lood C, Olsson ML, Izui S, et al. The IgG- specific endoglycosidase EndoS inhibits both cellular and complement- mediated autoimmune hemolysis. Blood 2010;115:5080e8. [40] van Timmeren MM, van der Veen BS, Stegeman CA, Petersen AH, Hellmark T, Collin M, et al. IgG glycan hydrolysis attenuates ANCA-mediated glomerulo- nephritis. J Am Soc Nephrol 2010;21:1103e14. [41] Schreiber A, Xiao H, Jennette JC, Schneider W, Luft FC, Kettritz R. C5a receptor mediates neutrophil activation and ANCA-induced glomerulonephritis. J Am Soc Nephrol 2009;20:289e98. [42] Wijngaarden S, van de Winkel JG, Jacobs KM, Bijlsma JW, Lafeber FP, van Roon JA. A shift in the balance of inhibitory and activating Fcgamma receptors on monocytes toward the inhibitory Fcgamma receptor IIb is associated with prevention of monocyte activation in rheumatoid arthritis. Arthritis Rheum 2004;50:3878e87. [43] Nimmerjahn F, Lux A, Albert H, Woigk M, Lehmann C, Dudziak D, et al. FcgammaRIV deletion reveals its central role for IgG2a and IgG2b activity in vivo. Proc Natl Acad Sci U S A 2010;107:19396e401. [44] Kasperkiewicz M, Nimmerjahn F, Wende S, Hirose M, Iwata H, Jonkman MF, et al. Genetic identification and functional validation of FcgammaRIV as key molecule in autoantibody-induced tissue injury. J Pathol; 2012. M. Hirose et al. / Journal of Autoimmunity 39 (2012) 304e314 314