ArticlePDF Available

Molecular and Therapeutic Characterization of Anti-ectodysplasin A Receptor (EDAR) Agonist Monoclonal Antibodies

Authors:

Abstract and Figures

The TNF family ligand ectodysplasin A (EDA) and its receptor EDAR are required for proper development of skin appendages such as hair, teeth and eccrine sweat glands. Loss of function mutations in the Eda gene cause X-linked hypohidrotic ectodermal dysplasia (XLHED), a condition that can be ameliorated in mice and dogs by timely administration of recombinant EDA. In this study, several agonist anti-EDAR monoclonal antibodies were generated that cross-react with the extracellular domains of human, dog, rat, mouse and chicken EDAR. Their half-life in adult mice was about 11 days. They induced tail hair and sweat gland formation when administered to newborn EDA-deficient Tabby mice, with EC50 of 0.1 to 0.7 mg/kg. Divalency was necessary and sufficient for this therapeutic activity. Only some antibodies were also agonists in an in vitro surrogate activity assay based on the activation of the apoptotic Fas pathway. Activity in this assay correlated with small dissociation constants. When administered in utero in mice or at birth in dogs, agonist antibodies reverted several ectodermal dysplasia features, including tooth morphology. These antibodies are therefore predicted to efficiently trigger EDAR signaling in many vertebrate species and will be particularly suited for long-term treatments.
Content may be subject to copyright.
Molecular and Therapeutic Characterization of
Anti-ectodysplasin A Receptor (EDAR) Agonist Monoclonal
Antibodies
*
S
Received for publication, June 3, 2011 Published, JBC Papers in Press, July 7, 2011, DOI 10.1074/jbc.M111.267997
Christine Kowalczyk
‡1
, Nathalie Dunkel
‡1,2
, Laure Willen
, Margret L. Casal
§
, Elizabeth A. Mauldin
§
, Olivier Gaide
,
Aubry Tardivel
, Giovanna Badic
‡3
, Anne-Lise Etter
4
, Manuel Favre
5
, Douglas M. Jefferson**
‡‡
, Denis J. Headon
§§
,
Ste´phane Demotz
¶¶6
, and Pascal Schneider
‡7
From the
Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland, the
§
School of Veterinary
Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6010, the
Department of Dermatology, University of
Geneva, CH-1211 Geneva, Switzerland,
Apoxis SA, CH-1004 Lausanne, Switzerland, **Cell Essential, Boston, Massachusetts 02116,
the
‡‡
Tufts University School of Medicine, Boston, Massachusetts 02111, the
§§
Roslin Institute and Royal (Dick) School of Veterinary
Studies, University of Edinburgh, Roslin EH25 9PS, Scotland, United Kingdom, and the
¶¶
Edimer Biotech, Ch de l’Eglise 7,
CH-1066 Epalinges, Switzerland
The TNF family ligand ectodysplasin A (EDA) and its receptor
EDAR are required for proper development of skin appendages
such as hair, teeth, and eccrine sweat glands. Loss of function
mutations in the Eda gene cause X-linked hypohidrotic ectoder-
mal dysplasia (XLHED), a condition that can be ameliorated in
mice and dogs by timely administration of recombinant EDA. In
this study, several agonist anti-EDAR monoclonal antibodies
were generated that cross-react with the extracellular domains
of human, dog, rat, mouse, and chicken EDAR. Their half-life in
adult mice was about 11 days. They induced tail hair and sweat
gland formation when administered to newborn EDA-deficient
Tabby mice, with an EC
50
of 0.1 to 0.7 mg/kg. Divalency was nec-
essary and sufficient for this therapeutic activity. Only some anti-
bodies were also agonists in an in vitro surrogate activity assay
based on the activation of the apoptotic Fas pathway. Activity in
this assay correlated with small dissociation constants. When
administered in utero in mice or at birth in dogs, agonist antibodies
reverted several ectodermal dysplasia features, including tooth
morphology. These antibodies are therefore predicted to effi-
ciently trigger EDAR signaling in many vertebrate species and will
be particularly suited for long term treatments.
The TNF family includes 19 ligands, most of which control
development, function and/or homeostasis of the immune sys-
tem (1). In this respect, ectodysplasin A (EDA)
8
is an exception
as it participates in ectodermal appendage formation (2). The
Eda gene on the X chromosome is transcribed as multiple splice
variants, only two of which code for the receptor-binding C-ter-
minal TNF homology domain. These two variants, generated
by splicing at an alternative donor site between exons 8 and 9,
code for 391- and 389-amino acid-long proteins called EDA1
and EDA2 (3). EDA1 binds EDAR, whereas EDA2 binds to
another receptor, XEDAR (3). The biology of EDA2 and
XEDAR is distinct from that of EDA1. Indeed, XEDAR-defi-
cient mice have no obvious ectodermal dysplasia phenotype,
whereas mice deficient in EDA, EDAR, or the signaling adaptor
protein EDARADD all display virtually indistinguishable ecto-
dermal dysplasia phenotypes, indicating the predominance of
the EDA1-EDAR axis in the development of skin-derived
appendages (4 8).
In humans, EDA1 loss of function mutations cause X-linked
hypohidrotic ectodermal dysplasia (XLHED), a rare condition
characterized by defective formation of teeth, hair, sweat glands
and other glands (6). Because of their insufficient number of
sweat glands, these patients are prone to hyperthermia. They
also frequently suffer from recurrent respiratory tract infec-
tions caused by abnormal mucus production in the airways.
Other problems are oligodontia, dry skin, and dry eyes (9–11).
EDA1 is a transmembrane type II protein with a furin con-
sensus cleavage site, a collagen-like domain, and a C-terminal
TNF homology domain, any of which when mutated can cause
XLHED (12). To be active, EDA must be processed and bind
EDAR through its trimeric C-terminal domain. The signaling
ability of EDA1 is re-enforced by its collagen domain that cross-
links individual EDA1 trimers (13). Interestingly, some EDA1
mutations can also cause selective tooth agenesis, a condition
characterized by no or very little involvement of other ectoder-
mal appendages (14). In these patients, EDA1 mutants retain
partial binding to EDAR, suggesting that tooth development is
particularly sensitive to “high quality” EDAR signals.
*This work was supported by grants from the Swiss National Science Foun-
dation (to P. S.) and by National Institutes of Health Grant RR02512 (to
M. L. C.). P. S., M. L. C., O. G., and S. D. are shareholders of EdimerPharma.
S
The on-line version of this article (available at http://www.jbc.org) contains
supplemental Figs. S1–S8.
The nucleotide sequence(s) reported in this paper has been submitted to the Gen-
BankTM/EBI Data Bank with accession number(s) JN099705 to JN099733.
1
Both authors contributed equally to this work.
2
Present address: Dept. of Oncology, Geneva University Hospital, CH-1211
Geneva, Switzerland.
3
Present address: Instrumat AG, Rueyre 116, CH-1020 Renens, Switzerland.
4
Present address: Celgene International, CH-2074 Marin, Switzerland.
5
Present address: Merck-Serono, CH-1809 Fenil-sur-Corsier, Switzerland.
6
Present address: Philip Morris Products SA, CH-2000 Neuchaˆtel, Switzerland.
7
To whom correspondence should be addressed. Tel.: 41-21-692-5709; Fax:
41-21-692-5705; E-mail: pascal.schneider@unil.ch.
8
The abbreviations used are: EDA, ectodysplasin A; EDAR, EDA receptor;
XLHED, X-linked hypohidrotic ectodermal dysplasia.
THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 35, pp. 30769–30779, September 2, 2011
© 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.
SEPTEMBER 2, 2011VOLUME 286• NUMBER 35 JOURNAL OF BIOLOGICAL CHEMISTRY 30769
by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from by guest on January 13, 2016http://www.jbc.org/Downloaded from
Transgenic expression of EDA1 in skin under the keratin 14
promoter results in a disheveled hair phenotype, hypertrophy
of sebaceous glands, and formation of supernumerary molars
or nipples (15). Transgenic EDA1 expression in the skin of
EDA-deficient Tabby mice corrected many of the ectodermal
dysplasia defects (16). The reverted phenotype was stable even
after shutdown of transgenic EDA1 expression in young adults,
suggesting that EDA1 plays a role in the formation but not in
the maintenance of skin appendages. Interruption of EDA1
expression, however, resulted in the normalization of seba-
ceous gland size (16). Similar conclusions were reached with an
alternative approach of protein replacement therapy, in which
EDA-deficient animals were exposed to a recombinant form of
EDA during development (17, 18). Taken together, these data
provide a proof of concept for protein replacement therapy in
young patients with XLHED.
In this study, we generated agonist anti-EDAR antibodies
that mimic the action of transgenic or recombinant EDA1 in
development. Most of these antibodies cross-react with EDAR
of mammals and birds and are active as monomeric, divalent
molecules. They corrected, among others, sweat glands, tra-
cheal glands, and tooth morphology in EDA-deficient mice and
were also active in EDA-deficient dogs. These mouse monoclo-
nal antibodies will be reagents of choice for long term experi-
ments in mice and pave the way for the development of thera-
peutic antibodies for use in XLHED or other EDAR-related
applications in humans.
EXPERIMENTAL PROCEDURES
Animals—Mice were handled according to Swiss Federal
Veterinary Office guidelines, under the authorization of the
Office Ve´te´rinaire Cantonal du Danton de Vaud (authorization
1370.3 to P. S.). White-bellied agouti B6CBAa A
wJ
/A-Eda
Ta
/J
Tabby mice (000314; The Jackson Laboratory) were bred as
Eda
Ta
/Eda
Ta
and Eda
Ta
/Y mutants or as /and /Y wild
type controls. EDAR-deficient OVE1B mice were as described
previously (5). EDA-deficient dogs (19) were cared for in
accordance with the principles outlined in the National Insti-
tutes of Health Guide for the Care and Use of Laboratory Ani-
mals and in the International Guiding Principles for Biomedical
Research Involving Animals.
Plasmids and Recombinant Proteins—Plasmids used in this
study were either previously published or derived from the pub-
lished plasmids by standard molecular biology techniques (sup-
plemental Fig. S1) (13, 20, 21). A fully human form of Fc-EDA1
was kindly provided by Dr. Neil Kirby (EdimerPharma, Boston).
hEDAR-Fc and mEDAR-Fc were produced and purified as
described previously (21).
Generation and Purification of Anti-EDAR Monoclonal
Antibodies—150
g of hEDAR-Fc or mEDAR-Fc (amino acid
residues 29–183 supplemental Fig. S1) were briefly sonicated
three times in 750
l of PBS/STIMUNE (1:1, v/v) (Cedi-diag-
nostics, Lelystad, The Netherlands). Female OVE1B mice were
immunized subcutaneously (base of tail, 200
l) with the anti-
gen preparation and boosted between days 10 and 14 with anti-
gen in PBS/STIMUNE (base of tail, 150
l). Mice positive for
anti-EDAR antibodies at day 30 were boosted with 150
gof
antigen in PBS at day 40 (base of tail). Three days later, lymph
node cells were fused with myeloma cells according to standard
procedures, grown in complete RPMI 1640 medium over a
feeder layer of mouse macrophages, and selected 24 h later
with hypoxanthine/aminopterin/thymidine-containing medium.
Supernatants of 96-well plates were tested by ELISA for antibody
secretion. Positive clones were subcloned twice by limiting dilu-
tion and then slowly adapted to medium without macrophages
and hypoxanthine/aminopterin/thymidine medium supplement.
Most hybridomas could then be progressively adapted to serum-
free Opti-MEM medium (Invitrogen). Antibodies were purified
from conditioned Opti-MEM supernatants by affinity chromatog-
raphy on protein G-Sepharose (GE Healthcare).
Transfections—HEK 293T cells were grown in DMEM, 10%
fetal calf serum and transfected by the calcium phosphate
method. Cells were grown for 7 days in serum-free Opti-MEM
medium (Invitrogen) for the production of EDAR-Fc trunca-
tion mutant fusion proteins or for 48 h in complete medium for
surface expression of receptors-TRAILR3 fusion proteins.
ELISA—For the detection of anti-EDAR antibodies, ELISA
plates were coated with hEDAR-Fc at 1
g/ml, blocked, and
revealed with anti-EDAR antibodies (adequately diluted serum
of EDAR immunized mice, hybridoma supernatants, or puri-
fied antibody) followed by a peroxidase-coupled goat anti-
mouse IgG (Jackson ImmunoResearch). For isotype determina-
tion, ELISA plates were coated with 1
g/ml of anti-EDAR
antibodies and revealed with peroxidase-coupled antibodies
against the heavy chain of mouse IgG1, IgG2a, or IgG2b (South-
ern Biotech). For epitope mapping, ELISA plates were coated
with an F(ab)
2
fragment of a goat anti-human Ig (Jackson
ImmunoResearch) to capture various EDAR-Fc constructs or a
control receptor (B cell maturation antigen-Fc) in cell superna-
tants. EDAR-Fc constructs and B cell maturation antigen-Fc
were revealed either with peroxidase-coupled donkey anti-hu-
man (HL) (Jackson ImmunoResearch) or with anti-EDAR
antibodies at 1
g/ml followed by peroxidase-coupled goat
anti-mouse IgG (Jackson ImmunoResearch) or with FLAG-
EDA1 or FLAG-BAFF followed by biotinylated anti-FLAG M2
antibody (Sigma) and peroxidase-coupled streptavidin.
SDS-PAGE, Western Blot, and Native Gel Electrophoresis
Anti-EDAR antibodies (10
g/lane) were analyzed by SDS-
PAGE under reducing conditions followed by Coomassie Blue
staining. Antibodies were also analyzed by native gel electro-
phoresis (4
g/lane) (Biomidi, Toulouse, France) and stained
with Amido Black according to manufacturer’s instructions,
except that the electrophoresis was performed for 1 h.
To test the ability of anti-EDAR antibodies to recognize
denatured EDAR, 2
g of bovine serum albumin, 20 ng of
hEDAR-Fc, and 20 ng of hFas-Fc were analyzed by SDS-
PAGE and Western blot under reducing (100 mMdithiothre-
itol) or nonreducing conditions, revealed with the various
anti-EDAR antibodies at 1
g/ml followed by peroxidase-
coupled anti-mouse antibody (1:10,000) and ECL reagent
(GE Healthcare).
Sequencing of Anti-EDAR Antibodies—RNA was extracted
from hybridoma cells with an RNeasy kit (Qiagen) according
to the manufacturer’s guidelines. cDNA was prepared by
reverse transcription with Ready-To-Go T-Primed first-
Strand kit (GE Healthcare). Variable sequences of the heavy
Agonist Anti-EDAR Antibodies
30770 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 •NUMBER 35 •SEPTEMBER 2, 2011
by guest on January 13, 2016http://www.jbc.org/Downloaded from
and light chains were amplified by PCR as described previ-
ously (22). PCR products were sequenced on both strands.
Sequences were analyzed for gene usage using the IMGT
sequence alignment software.
FACS Analyses—293T cells co-transfected with enhanced
GFP, and receptor-glycosylphosphatidylinositol expression
plasmids were stained with Fc-EDA1 or Fc-EDA2 essentially as
described before (20) or with anti-EDAR antibodies at 4
g/ml
followed by phycoerythrin-coupled anti-mouse secondary anti-
body. Following staining, cells were analyzed using a FACScan
flow cytometer (BD Biosciences) and FlowJo software (TreeStar,
Ashland, OR).
Fab Generation and Affinity Determination by Surface Plas-
mon Resonance—An IgG1 Fab and F(ab)
2
preparation kit was
used according to the manufacturer’s instructions (Pierce).
Briefly, purified anti-EDAR antibodies were digested for 72 h at
37 °C with immobilized ficin. Fc fragments and undigested
antibodies were removed by chromatography on protein A.
The flow-through, containing the Fab and F(ab)
2
fragments,
was concentrated and applied onto a Superdex-200 gel perme-
ation chromatography column eluted in PBS. Absorbance was
recorded at 280 nm.
For surface plasmon resonance, human EDAR-Fc was cap-
tured on anti-human IgG Fc-derivatized CM5 chips in a Bia-
core T100 apparatus (GE Healthcare). Fab solutions of anti-
EDAR antibodies at the indicated concentration in PBS were
applied for 90 s at 50
l/min and subsequently washed with
buffer. All curve fittings were performed assuming a 1:1 binding
model, although two antibodies had a biphasic dissociation that
did not fit the 1:1 model.
In Vitro Cytotoxicity Assays—Fas-deficient hEDAR:Fas Jur-
kat cells have been described before, and mEDAR:Fas-express-
ing cells were obtained by retroviral infection according to the
same protocol (13). The cytotoxicity assay using EDAR:Fas Jur-
kat cells was performed as described for FasL on Jurkat cells
(23).
Injections in EDA-deficient AnimalsTabby pups were
labeled by puncture of a footpad with a 30-gauge needle dipped
in china ink. Intraperitoneal injections with anti-EDAR anti-
bodies or Fc-EDA1 were performed within 24 h after birth with
a maximal volume of 15
l, using 0.5-ml U-100 insulin syringes
(BD Biosciences). Examination and photography of tail hairs
were performed at days 20–22 post-injection. Pregnant Tabby
mice were treated intravenously at days 13 and 20 (E13/E20) or
9 and 17 (E9/E17) of gestation with 400
g of anti-EDAR anti-
body (antibody 3). Offspring were analyzed at 6 months of age,
essentially as described previously (18). Age-matched wild type
and EDA-deficient Tabby mice were similarly analyzed for
comparison. Tracheal glands were detected by Alcian blue
staining (24). Three dogs affected with X-linked ectodermal
FIGURE 1. Purity of protein-G purified anti-EDAR monoclonal antibodies.
Anti-EDAR monoclonal antibodies were purified by protein G affinity chroma-
tography from culture supernatants in serum-free medium (antibodies 1– 4,
6 –10, and 12–14) or in serum-containing medium (antibodies 5, 11, and 15).
A, SDS-PAGE analysis and Coomassie Blue staining of 10
g of antibody per
lane under reducing conditions. Migration positions of molecular mass stan-
dards (in kDa) are shown. B, native gel electrophoresis of 4
g of anti-EDAR
antibodies per lane, stained with Amido Black.
TABLE 1
Characteristics of anti-EDAR monoclonal antibodies
IGHV and IGLV indicate immunoglobulin heavy and light chain variable region genes likely used in the antibody. hu is human EDAR-Fc; mu is mouse EDAR-Fc.III
indicates EDAR-Fc containing CRD1 and CRD2 of human EDAR. I is EDAR-Fc containing CRD1 only. Full is EDAR-Fc containing the full extracellular domain of EDAR.
k
a
is the association constant. k
d
is the dissociation constant. K
D
is the affinity of monomeric Fab to EDAR-Fc. EC
50
tail hair indicates the dose of antibody required to get
half-maximal tail hair reversion score when administered intraperitoneally in newborn Tabby mice. EC
50
, mEDAR:Fas or hEDAR:Fas means dose of antibody required to
kill half of the EDAR:Fas-expressing Jurkat cells.
Anti-EDAR IGHV gene IGLV gene Antigen Isotype Epitope k
a
10
5
k
d
10
4
K
D
EC
50
Tail hair mEDAR:Fas hEDAR:Fas
M
1
s
1
s
1
nMmg/kg ng/ml ng/ml
1 5–17 10–96 hu IgG1 I II 6.79 3.69 0.54 0.125 10 200
2 1–69 1–117 hu IgG1 I II 13.2 13.3 1.00 0.42 4000 4000
3 5–17 10–96 hu IgG1 I II ND ND ND 0.18 30 100
4 ND 1–117 hu IgG1 I II 4.50 13.6 3.01 0.7 4000 4000
5 2–2 4–91 hu IgG2b I ND ND ND 0.25 4000 4000
6 1S135 1–110 hu IgG2a I 3.81 6.02 1.58 0.25 4000 4000
7 7–3 4–577 hu IgG1 I II 2.92 96.6 33.1 0.3 4000 4000
8 5–17 10–96 mu IgG1 I II 8.12 3.86 0.48 0.5 100 4000
9 1–63 1–117 mu IgG1 IIIIII 1.89 8.44 4.46 0.42 4000 4000
10 1–39 4–55 hu IgG1 I II 0.35 2.38 6.78 0.125 10 50
11 1–39 10–94 hu IgG1 I II ND ND ND 3.3 4000 4000
12 1–14 17–12 mu IgG1 I 0.41 6.66 16.3 0.35 10 5
13 1S135 1–110 hu IgG1 I 1.83 18.7 10.2 0.42 4000 4000
14 1–42 10–94 mu IgG1 I II 1.90 74.8 39.3 0.3 4000 4000
15 1–63 12–44 hu IgG1 I II ND ND ND ND ND ND
Agonist Anti-EDAR Antibodies
SEPTEMBER 2, 2011VOLUME 286• NUMBER 35 JOURNAL OF BIOLOGICAL CHEMISTRY 30771
by guest on January 13, 2016http://www.jbc.org/Downloaded from
dysplasia were administered agonist anti-EDAR antibody 3 in
the jugular vein at 2 days of life (n2; 10 mg/kg) or at 14 days
of life (n1; 7 mg/kg). The analysis was performed essentially
as described previously (17, 25). In particular, the dogs were
monitored daily for adverse reactions, overall health, and spe-
cific ocular and respiratory diseases. Complete blood cell
counts and serum biochemistry screens were evaluated within
2–7 days after injection with agonist anti-EDAR antibody. Den-
tal radiographs were obtained when the dogs were adults, when
about 1 year old. Shirmer tear testing was performed at
6-month intervals. Complete necropsies were performed
between 1.6 and 2.4 years of age. Tissues were fixed in 10%
neutral buffered formalin, routinely processed, sectioned at 5
m, and stained with hematoxylin and eosin.
RESULTS
Generation and Screening of Agonist Anti-EDAR Antibodies
To obtain cross-reacting antibodies against conserved EDAR
regions, EDAR-deficient mice were immunized with either
human or mouse EDAR-Fc fusion proteins. The EDAR-defi-
cient mouse strains Downless and Sleek have loss-of-function
mutations in the extracellular or intracellular domains of
EDAR, respectively, but still express the protein. We therefore
immunized OVE1B mice in which the Edar gene is completely
FIGURE 2. Epitope mapping of anti-EDAR monoclonal antibodies. A, schematic linear representation of human EDAR showing the position of cysteine
residues (thin horizontal lines), the putative N-linked glycosylation site (thick horizontal line, N), and the six structural modules (rectangles with rounded corners)
composing the three cysteine-rich domains (CRD1, CRD2, and CRD3). The transmembrane domain (TMD), signal peptide (Leader), stalk, and intracellular
domain (ID) are also shown. Amino acid numbers at the junctions of interest are indicated. The arrowhead indicates the predicted cleavage site of the signal
peptide. The scheme is drawn to scale, except for the intracellular domain. Ctrl, control. B, the indicated EDAR-Fc constructs or B cell maturation antigen-Fc
(BCMA-Fc) control were captured in an ELISA plate and revealed with the indicated anti-EDAR antibodies, with an anti-human IgG to control efficient capture
of the various EDAR-Fc proteins, or with FLAG-EDA1 or FLAG-BAFF as controls. C, bovine serum albumin (B; BSA, 2
g), hEDAR-Fc (E, 20 ng), and hFas-Fc (F,20
ng) were resolved by SDS-PAGE under reducing or nonreducing conditions, transferred onto nitrocellulose, and probed with anti-EDAR1–15.
Agonist Anti-EDAR Antibodies
30772 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 •NUMBER 35 •SEPTEMBER 2, 2011
by guest on January 13, 2016http://www.jbc.org/Downloaded from
deleted by random genomic integration of an unrelated trans-
gene (5, 26). Hybridoma supernatants with anti-EDAR reactiv-
ity were screened for agonist activity using two complementary
tests. In the first one, surrogate reporter cell lines stably
expressing hEDAR:Fas or mEDAR:Fas fusion proteins were
used. EDAR activation in these reporter cells leads to apoptotic
cell death by activation of the Fas pathway (13). In the second
assay, hybridoma supernatants were administered intraperito-
neally to newborn, EDA-deficient Tabby pups (18). Tabby mice
completely lack tail hairs, and the agonist activity of antibodies
can therefore be measured by the induction of tail hair, pro-
vided that antibodies recognize and activate mouse EDAR. Sev-
eral hybridoma secreted agonist antibodies with in vivo activity,
but only a few were also active in the cell-based in vitro assay
(data not shown). Selected hybridoma were subcloned and
adapted for growth in serum-free medium from which antibod-
ies were purified (Fig. 1A). The monoclonal nature of these
antibodies was further confirmed by their sharp migration in
native protein electrophoresis (Fig. 1B).
Agonist Anti-EDAR Antibodies Have Varied but Relatively
Limited Sequence Characteristics—Variable regions of heavy
and light antibody chains were amplified by RT-PCR and
sequenced for several hybridoma. A number of different vari-
able region genes were identified for both the heavy and light
chains (Table 1), but some of them were shared by two or three
hybridoma, usually with different somatic mutations. Interest-
ingly, antibodies with identical heavy and light variable genes
sharing greater than 90% sequence identity were obtained from
different mice immunized with mouse (antibody 8) or human
EDAR (antibodies 1 and 3) (Table 1 and supplemental Fig. S2).
Thus, different variable genes can be used to generate agonist
anti-EDAR antibodies, but the gene repertoire must be limited
as similar antibodies were found two or three times in the rela-
tively limited panel that we have analyzed.
Agonist Antibodies Recognize at Least Three Different
Epitopes in EDAR—EDAR contains three cysteine-rich do-
mains in its extracellular region, plus a stalk sequence (Fig. 2A).
We used EDAR constructs containing these four regions alone
or in combinations to roughly characterize the epitopes recog-
nized by the antibodies. Some antibodies recognized CRD1
alone, others recognized CRD1 and -2 together, and one
reacted with CRD1–3, but none bound to receptors lacking
CRD1 (Fig. 2Band Table 1). All antibodies recognized recom-
binant EDAR by Western blot under nonreducing conditions,
but only three reacted with reduced EDAR (Fig. 2C). These
three antibodies had their epitopes localized in CRD1. It is pos-
sible that the presence of CRD1 is required for correct folding of
EDAR. Abnormal disulfide bridge formation in the absence of
FIGURE 3. Cross-species specificity of anti-EDAR antibodies. Receptors fused to the glycosylphosphatidylinositol anchor of TRAILR3 were expressed in 293T
cells together with an enhanced GFP tracer (xaxis) and stained with Fc-EDA1, Fc-EDA2, or with anti-EDAR antibodies (yaxis). Receptor expression was
confirmed by staining with an antibody directed against the C-terminal portion of TRAIL-R3 present in all constructs (mAb572). Both scattergram axes show
fluorescence intensity on a logarithmic scale (10
0
–10
4
).
Agonist Anti-EDAR Antibodies
SEPTEMBER 2, 2011VOLUME 286• NUMBER 35 JOURNAL OF BIOLOGICAL CHEMISTRY 30773
by guest on January 13, 2016http://www.jbc.org/Downloaded from
CRD1 would explain why antibodies did not recognize con-
structs containing CRD2 and CRD3. Taken together, these
results indicate that agonist anti-EDAR antibodies can recog-
nize at least three different EDAR epitopes located in CRD1 and
probably CRD2 and CRD3.
Agonist Anti-EDAR Antibodies Cross-react with EDAR of
Various Mammals and Birds—Most anti-EDAR antibodies
cross-reacted with human, dog, rat, mouse, and chicken
EDAR when these were expressed as glycosylphosphatidyli-
nositol-anchored molecules in 293T cells (Fig. 3). Antibody 5
only reacted minimally with chicken EDAR, whereas anti-
body 15 that had been selected on the basis of its specificity
for human EDAR rather than for its agonist activity recog-
nized human and dog EDAR, but not rat, mouse and chicken
EDAR (Fig. 3).
Binding Characteristics of Anti-EDAR Antibodies to EDAR
The affinity of 11 agonist antibodies to human EDAR was
determined by surface plasmon resonance. For this purpose,
monomeric Fab fragments were generated by ficin digestion
and size exclusion chromatography (Fig. 4A). Affinities var-
ied from 0.5 to 40 nM, and differences were also observed in
the association and dissociation constants (Fig. 4Band Table
1). The dissociation kinetics of two antibodies (7 and 14)
were biphasic, showing first a rapid dissociation followed by
a slower dissociation, but as these antibodies had no remark-
able agonist activity, this was not analyzed further.
Comparison of the Activity of Agonist Anti-EDAR Antibodies
in Vitro and in Vivo—As we have not yet been able to identify a
simple and quantitative assay to characterize EDAR agonists
using EDAR’s own signaling pathway in vitro, we used a surro-
gate reporter assay in which Fas-sensitive cells were transfected
with the extracellular domain of human or mouse EDAR fused
to the intracellular portion of Fas. Binding of an active recom-
binant EDA1 (Fc-EDA1) to these cells induces cell death by
activation of the pro-apoptotic Fas pathway (13). Interestingly,
only some of the antibodies (1, 3, 8, 10, and 12) (Fig. 5A) killed
mEDAR:Fas-expressing cells, and even fewer killed hEDAR:Fas
expressing cells (1, 3, 10, and 12) (Fig. 5Band Table 1). In all
cases, antibodies were less active than Fc-EDA1 by 1–2 orders
of magnitude. The picture was different in an in vivo assay,
where newborn Tabby mice were treated with antibodies on the
day of birth. In this case, all antibodies rescued tail hair forma-
tion in a dose-dependent manner and with similar EC
50
values
of 0.1 to 0.7 mg/kg (Fig. 6). Only one of the antibodies (antibody
11) seemed less active, with an EC
50
value of about 3 mg/kg.
Functional sweat glands were induced by the treatment with
similar EC
50
as for tail hair (data not shown). The half-life of
two antibodies (1 and 3) was determined in adult Tabby mice
and found to be 10.5 and 11 days, respectively (supplemental
Fig. S3).
Divalent Monomeric Agonist Antibodies Are Active in Vivo
We have shown previously that cross-linked EDA1 containing
more than one trimer in a single molecule are better agonists
than trimeric EDA1 (13). We therefore wondered whether the
in vivo activity of agonist anti-EDAR antibodies was due to
monomeric antibodies or to aggregates thereof. The activity of
a monomeric (divalent) antibody purified by size exclusion
chromatography (Fig. 6, antibody 1) was, however, very similar
to that of the total preparation (data not shown), and compared
favorably (EC
50
0.1 mg/kg) with recombinant Fc-EDA1 (EC
50
0.05 mg/kg) (Fig. 6). In addition, the F(ab)
2
fragment, but not
the Fab fragment, was active in vivo when administered to new-
born pups on the day of birth (supplemental Fig. S4). When the
F(ab)
2
was administered later at day 3 post-birth, one of the
latest time points where ventral tail hair can be induced, its
action could be inhibited by an excess of the Fab fragment,
ruling out that the lack of agonist activity of the Fab would be
due only to a shorter half-life in vivo (supplemental Fig. S4). We
FIGURE 4. Generation and binding characteristics of anti-EDAR Fab fragments. A, Superdex-200 gel permeation chromatography elution profiles of intact
and ficin-digested anti-EDAR antibodies. In the digested antibodies, Fc fragments and undigested antibodies were first removed by chromatography on
protein A. Peak identities are indicated. B, Fab fragments at the indicated concentrations were analyzed by surface plasmon resonance onto immobilized
hEDAR-Fc. Fab solutions were applied for 90 s, and subsequently washed with buffer. Results for three antibodies with distinct binding characteristics are
shown.
Agonist Anti-EDAR Antibodies
30774 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 •NUMBER 35 •SEPTEMBER 2, 2011
by guest on January 13, 2016http://www.jbc.org/Downloaded from
conclude from these observations that a divalent agonist anti-
EDAR antibody is both sufficient and necessary to exert activity
in Tabby mice.
Effective Treatment of EDA-deficient Mice with Agonist Anti-
EDAR Antibodies—Some patients with partially inactivating
EDA mutations have teeth defects but otherwise normal skin
appendages, suggesting that tooth formation may require more
stringent EDAR signals than other skin appendages for proper
development (14). To test the effect of agonist antibodies on
tooth development, pregnant Tabby mothers were treated dur-
ing pregnancy so that the antibody could be transferred to
embryos by the trans-placental antibody transport system.
Mice exposed to agonist anti-EDAR during development not
only had tail hairs and functional sweat glands, but also hair
behind the ears, mucus-secreting glands in the trachea, and a
normalized eye appearance (Fig. 7, A–G, and supplemental Fig.
S5). In addition, molars of treated mice were reverted and
almost indistinguishable from those of wild type animals (Fig. 7,
Hand I, and supplemental Fig. S5). The effect was long lasting,
as a similarly treated mouse was still reverted after more than 2
years (supplemental Fig. S6). Taken together, these results indi-
cate that the two agonists anti-EDAR antibodies tested in this
application (antibodies 1 and 3, Fig. 7, supplemental Fig. S4, and
data not shown) revert the ectodermal dysplasia phenotypes
that we have looked at in Tabby mice, including tooth
morphology.
Activity of Agonist Anti-EDAR Antibodies in EDA-deficient
Dogs—Agonist anti-EDAR antibodies recognize EDAR of dif-
ferent species (Fig. 3). To test whether the observed cross-spe-
cies reactivity also holds true for the agonist activity, anti-EDAR
antibody 3 was administered intravenously to three EDA-defi-
cient dogs at either 2 days of life (n2, 10 mg/kg) or at 14 days
of life (n1, 7 mg/kg). None of these dogs showed adverse
reactions upon injection. Dentition was corrected not only in
FIGURE 5. In vitro activity of anti-EDAR antibodies in a surrogate reporter assay. A, anti-EDAR antibodies and Fc-EDA1 were tested for their capacity to
induce apoptosis of mEDAR:Fas-expressing Jurkat cells. After overnight culture, cell viability was determined by a cell viability assay. EC
50
are indicated as
dotted lines. Anti-EDAR antibody number (or Fc-EDA1) for each curve is indicated to the right of each panel. B, as above, except that hEDAR:Fas expressing cells
were used.
FIGURE 6. Therapeutic doses of the anti-EDAR antibodies in newborn Tabby mice. Newborn Tabby mice were injected intraperitoneally during the first 24 h
of life with graded doses of anti-EDAR antibodies, an irrelevant mouse IgG1 (Aprily 5) or Fc-EDA1. Four to 6 weeks later (antibody 2– 4) or 3 weeks later
(antibodies 1, 4 –14, and Aprily 5), hair density on the tail was scored according to the criteria shown in the inset. The anti-EDAR antibody 1 used in this
experiment was the monomer peak obtained by gel filtration (see Fig. 4).
Agonist Anti-EDAR Antibodies
SEPTEMBER 2, 2011VOLUME 286• NUMBER 35 JOURNAL OF BIOLOGICAL CHEMISTRY 30775
by guest on January 13, 2016http://www.jbc.org/Downloaded from
EDA-deficient dogs that were treated at 2 days of life but also in
the affected treated at 14 days of life, although the latter still
lacked premolars, accounting for the decreased number of
teeth (Fig. 8 and Table 2). Interestingly, the premolars and
molars in dogs treated at day 2 of life had a more normal
appearance than in those dogs treated with Fc-EDA (17).
Lacrimation was improved in treated dogs except in one
treated at day 2 of age (Table 2). The correction of glands in
trachea, bronchi, and esophagus appeared, however, to be
dependent on the age at which the dogs were treated, i.e.
treatment administered earlier in life had a bigger impact on
gland development (Fig. 8 and Table 2). It is noteworthy that,
regardless of the extent of the phenotypic reversion, none of
the treated dogs suffered from pneumonia or other airway
diseases that are common in untreated EDA-deficient dogs
or from dry eye condition (keratoconjunctivitis sicca) that
affect all XLHED dogs.
DISCUSSION
Deficiency in the TNF family ligand EDA leads to ectoder-
mal dysplasia, even if the receptor EDAR remains fully func-
tional. The development of EDAR agonists are thus of inter-
est for applications in the treatment of XLHED. EDAR is
relatively well conserved across species, with only 4 amino
acid differences between human and dog, 10 between human
and mouse, and 13 between human and chicken in the 154-
amino acid-long mature extracellular domain (supplemental
Fig. S7). To increase the likelihood of getting cross-reactive
antibodies, EDAR-deficient mice were used to generate
monoclonal antibodies. The approach proved successful as
13 of the 15 anti-EDAR antibodies analyzed recognized
EDAR from human, dog, rat, mouse, and chicken. These
antibodies are therefore likely to cross-react with EDARs of
all mammals and many other vertebrates. Anti-EDAR15 dif-
ferentiated human and dog from rat and mouse EDARs
whose primary sequence only diverge in CRD1 (supplemen-
tal Fig. S7), implying that part of its epitope is in CRD1.
Anti-EDAR9 recognized CRD1–3 of EDAR but none of the
cysteine-rich domains taken individually or in pairs, despite
the fact that these fragments were overlapping and supposed
to respect structural elements of the receptor. These results
indicate that EDAR may fold on itself to create a conforma-
tional epitope with regions that are distant in the primary
FIGURE 7. Anti-EDAR antibody reverts many ectodermal dysplasia phenotypes in EDA-deficient mice. Pregnant Tabby mice were treated intravenously
at days 13 and 20 (E13/E20) of gestation with anti-EDAR antibody 3 at 16 mg/kg. Offspring were analyzed at 6 months of age. Age-matched wild type and
EDA-deficient Tabby mice were similarly analyzed for comparison. A, tail phenotype. B, transversal sections of tail skin showing the presence of hair follicles,
stained with hematoxylin and eosin. C, starch iodine sweat tests. D, sections of foot pads showing the presence of glandular structures of sweat glands
(arrowheads), stained with hematoxylin and eosin. E, top view of the retro-auricular region showing the presence of hair behind the ears. F, sections of the
trachea stained with Alcian blue to reveal mucus-secreting glands (arrowheads). G, eye phenotype showing reversion of the thickened eyelid margin and
narrow eyelid opening. Hand I, pictures of the jaw carrying the upper and lower molars. Antibody treated and wild type jaws carry larger teeth with a normal
pattern of cusps on their surfaces.
Agonist Anti-EDAR Antibodies
30776 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 •NUMBER 35 •SEPTEMBER 2, 2011
by guest on January 13, 2016http://www.jbc.org/Downloaded from
sequence. In any case, results indicate that agonist anti-
EDAR antibodies can recognize at least three distinct
epitopes in EDAR.
Anti-EDAR antibodies were screened in newborn Tabby
mice, a highly relevant in vivo assay whose main limitation is to
detect only antibodies cross-reacting with mouse EDAR. More
than half of the 46 hybridoma supernatants tested were active in
this assay, indicating that agonist antibodies can readily be
obtained with the procedure used. The success rate of detection
was lower using the EDAR:Fas reporter cell lines that signal
cell death in an oligomerization-sensitive manner, probably
because these cell-based assays make use of a different intracel-
lular signaling pathway. When combined, these assays discrim-
inated two classes of agonist anti-EDAR antibodies, with or
without in vitro activity. Interestingly, the in vitro activity cor-
related relatively well with low antibody dissociation constants
but not with association constants or affinities (supplemental
Fig. S8). The EDAR:Fas reporter cells were previously shown
to discriminate recombinant WT EDA1 and EDA1 with the
V365A mutation identified in a family with selective tooth
agenesis, despite the fact that these two ligands bind EDAR-Fc
almost equally well (14). This led to the hypothesis that forma-
tion of teeth may require higher quality EDAR signals than
those needed for hair or gland formation. Thus, it is possible
that agonist antibodies with activity in vitro may also be the best
ones to correct tooth defects associated with EDA deficiency. It
will be interesting to experimentally test this hypothesis in the
future.
The plethora of anti-EDAR agonist antibodies obtained,
most of which were of the IgG1 isotype, was surprising.
Indeed, there are indications that agonist anti-Fas antibodies
(Fas is another TNFR family member) need to be oligomer-
ized to be active. For example, the CH11 IgM monoclonal
antibody directed against human Fas was obtained by immu-
nization of mice with membranes of FS-7 human fibroblasts
(27). When an IgG1 recognizing the exact same epitope
(mAb ZB4) or a divalent F(ab)
2
of CH11 was used, there was
no agonist activity (28). A second example of an agonist
FIGURE 8. Anti-EDAR antibody ameliorates dentition and presence of glands in EDA-deficient dogs. An EDA-deficient dog was treated at day 2 of
life with a single dose of anti-EDAR (antibody 3) at 10 mg/kg, and analyzed 1.6 years later in comparison with a wild type and with an affected dog. Aand
B, front and side views of the jaws. Cand D, hematoxylin and eosin-stained tissue sections of the trachea and bronchi. Glandular tissues are indicated
with arrowheads.
TABLE 2
Summary of clinical and pathological findings in untreated XLHED dogs and XHLED dogs treated with anti-EDAR antibody 3
Dog number Treatment protocol
Age at
necropsy
Appearance of teeth
(number)
Tracheal
glands
Bronchial
glands
Esophageal
glands Tear production
%
Wild type (n5) 1–3 years Normal (41.6 0.9) ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ 97 14
XLHED (n6) 1–3 years Abnormal (18.0 2.9) None None None 68 20
E237 10 mg/kg on day 2 1.7 years Greatly improved (40) ⫹⫹⫹ ⫹⫹⫹ ⫹⫹⫹ 70.0 4.1
E241 10 mg/kg on day 2 1.6 years Greatly improved (41) ⫹⫹ ⫹ ⫹⫹ 90.0 7.1
E222 7 mg/kg on day 14 2.4 years Improved (30) ⫹⫺ ⫹ 80.0 27.1
Agonist Anti-EDAR Antibodies
SEPTEMBER 2, 2011VOLUME 286• NUMBER 35 JOURNAL OF BIOLOGICAL CHEMISTRY 30777
by guest on January 13, 2016http://www.jbc.org/Downloaded from
monoclonal antibody directed against human Fas is APO-1,
which was obtained by immunizing mice with plasma mem-
branes of the human SKW 6.4 cell line (29). This antibody is
an IgG3. Upon isotype switch, it loses its agonist activity (30),
a result that was tentatively explained by the propensity of
IgG3 to self-aggregate. As immunoglobulin preparations
often contain low amounts of high molecular weight anti-
body aggregates (31), we wondered whether the agonist
activity of anti-EDAR antibodies could be due to aggregates.
This was not the case, however, as monomeric antibodies
had a similar specific activity in vivo as the total preparation.
In addition, a purified F(ab)
2
fragment was agonist in vivo,
whereas the Fab fragment was not. We conclude that diva-
lency is necessary and sufficient for anti-EDAR agonist anti-
bodies to exert their activity.
We have shown previously that the collagen domain of
EDA oligomerizes the trimeric TNF homology domain of
EDA1 into higher order structures with concomitant gain
of activity (13). Similarly, Fc-EDA1 that assembles as a hexamer
(containing two EDA1 trimers) is a highly active molecule.
One interpretation is that multiple EDAR molecules must be
recruited within the same complex to deliver robust intra-
cellular signals, but this is in apparent contradiction with the
observation that divalent antibodies are good agonists. A
first hypothesis to reconcile these observations is that EDAR
may pre-exist as inactive complexes before ligand binding, as
shown previously for Fas (32). Binding of a ligand may
change the conformation of the complex to render it signal-
ing-competent and bring together two such complexes to
initiate signaling. Divalent agonist antibodies may mimic
hexavalent ligands by inducing the conformational change
by binding one receptor in the pre-assembled complex and
recruiting and activating a second complex on its second
arm. In a second hypothesis, binding of the ligand or the
antibody at an appropriate site of EDAR is sufficient to
render the receptor signaling-competent. Assembly of the
signaling complex may be relatively slow and reversible if
the agonist detaches prematurely from the receptor. In this
model, efficient signaling could be obtained either with
divalent reagents with low dissociation constants or with
ligands that compensate relatively high dissociation con-
stants by multivalency. Whatever their mechanism of action,
Fc-EDA1 and agonist anti-EDAR antibodies are in practice
excellent agonists to cure animal models of XLHED, in-
cluding their teeth defects. Because of their long half-life
in vivo, agonist anti-EDAR antibodies will prove useful
reagents for long term experiments, especially in mice where
these mouse antibodies should elicit minimal neutralizing
immune responses.
Finally, it is noteworthy that keratoconjunctivitis sicca (dry
eye) that affects all untreated EDA-deficient dogs is believed to
be caused by decreased tear production. Tear production
improved significantly in two of the treated dogs but not in a
third one (E237). Nevertheless, none of the dogs treated in this
study with agonist anti-EDAR antibody required therapy for
dry eye or any other ocular disorders. These findings suggest
that decreased tear production is not the only factor causing dry
eyes but that other structures such as Meibomian glands that
lubricate the eye, or the composition of the lipids therein, also
play an important role.
Acknowledgments—We thank Christophe Maier (Edimer Biotech,
Epalinges) for invaluable input in the creation of Edimer; Jeff Behrens
and Neil Kirby for their motivating discussions, support, and for the
gift of Fc-EDA; Ste´phane Germain for assistance in early phases of
mice work; Olivier Donze´ for expert help in the generation of the anti-
bodies; John Lewis for dental pictures of the dogs; veterinary techni-
cians and students of the University of Pennsylvania for the expert
care of the dogs; and Ju¨rg Tschopp for continuous support and helpful
discussions.
REFERENCES
1. Aggarwal, B. B. (2003) Nat. Rev. Immunol. 3, 745–756
2. Mikkola, M. L. (2008) Cytokine Growth Factor Rev. 19, 219 –230
3. Yan, M., Wang, L. C., Hymowitz, S. G., Schilbach, S., Lee, J., Goddard, A.,
de Vos, A. M., Gao, W. Q., and Dixit, V. M. (2000) Science 290, 523–527
4. Headon, D. J., Emmal, S. A., Ferguson, B. M., Tucker, A. S., Justice,
M. J., Sharpe, P. T., Zonana, J., and Overbeek, P. A. (2001) Nature 414,
913–916
5. Headon, D. J., and Overbeek, P. A. (1999) Nat. Genet. 22, 370 –374
6. Monreal, A. W., Zonana, J., and Ferguson, B. (1998) Am. J. Hum. Genet. 63,
380–389
7. Newton, K., French, D. M., Yan, M., Frantz, G. D., and Dixit, V. M. (2004)
Mol. Cell. Biol. 24, 1608–1613
8. Srivastava, A. K., Pispa, J., Hartung, A. J., Du, Y., Ezer, S., Jenks, T., Shi-
mada, T., Pekkanen, M., Mikkola, M. L., Ko, M. S., Thesleff, I., Kere, J., and
Schlessinger, D. (1997) Proc. Natl. Acad. Sci. U.S.A. 94, 13069–13074
9. Nakata, M., Koshiba, H., Eto, K., and Nance, W. E. (1980) Am. J. Hum.
Genet. 32, 908–919
10. Pinheiro, M., and Freire-Maia, N. (1979) Am. J. Med. Genet. 4, 113–122
11. Zankl, A., Addor, M. C., Cousin, P., Gaide, A. C., Gudinchet, F., and Schor-
deret, D. F. (2001) Eur. J. Pediatr. 160, 296–299
12. Schneider, P., Street, S. L., Gaide, O., Hertig, S., Tardivel, A., Tschopp, J.,
Runkel, L., Alevizopoulos, K., Ferguson, B. M., and Zonana, J. (2001) J. Biol.
Chem. 276, 18819–18827
13. Swee, L. K., Ingold-Salamin, K., Tardivel, A., Willen, L., Gaide, O., Favre,
M., Demotz, S., Mikkola, M., and Schneider, P. (2009) J. Biol. Chem. 284,
27567–27576
14. Mues, G., Tardivel, A., Willen, L., Kapadia, H., Seaman, R., Frazier-Bow-
ers, S., Schneider, P., and D’Souza, R. N. (2010) Eur. J. Hum. Genet. 18,
19–25
15. Mustonen, T., Pispa, J., Mikkola, M. L., Pummila, M., Kangas, A. T., Pak-
kasja¨rvi, L., Jaatinen, R., and Thesleff, I. (2003) Dev. Biol. 259, 123–136
16. Cui, C. Y., Durmowicz, M., Ottolenghi, C., Hashimoto, T., Griggs, B.,
Srivastava, A. K., and Schlessinger, D. (2003) Hum. Mol. Genet. 12,
2931–2940
17. Casal, M. L., Lewis, J. R., Mauldin, E. A., Tardivel, A., Ingold, K., Favre, M.,
Paradies, F., Demotz, S., Gaide, O., and Schneider, P. (2007) Am. J. Hum.
Genet. 81, 1050–1056
18. Gaide, O., and Schneider, P. (2003) Nat. Med. 9, 614 618
19. Casal, M. L., Scheidt, J. L., Rhodes, J. L., Henthorn, P. S., and Werner, P.
(2005) Mamm. Genome 16, 524–531
20. Bossen, C., Ingold, K., Tardivel, A., Bodmer, J. L., Gaide, O., Hertig, S.,
Ambrose, C., Tschopp, J., and Schneider, P. (2006) J. Biol. Chem. 281,
13964–13971
21. Schneider, P. (2000) Methods Enzymol. 322, 325–345
22. Wang, Z., Raifu, M., Howard, M., Smith, L., Hansen, D., Goldsby, R., and
Ratner, D. (2000) J. Immunol. Methods 233, 167–177
23. Schneider, P., Holler, N., Bodmer, J. L., Hahne, M., Frei, K., Fontana, A.,
and Tschopp, J. (1998) J. Exp. Med. 187, 1205–1213
24. Rawlins, E. L., and Hogan, B. L. (2005) Dev. Dyn. 233, 1378 –1385
25. Mauldin, E. A., Gaide, O., Schneider, P., and Casal, M. L. (2009) Am. J.
Med. Genet. A 149A, 2045–2049
Agonist Anti-EDAR Antibodies
30778 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 •NUMBER 35 •SEPTEMBER 2, 2011
by guest on January 13, 2016http://www.jbc.org/Downloaded from
26. Shawlot, W., Siciliano, M. J., Stallings, R. L., and Overbeek, P. A. (1989)
Mol. Biol. Med. 6, 299–307
27. Yonehara, S., Ishii, A., and Yonehara, M. (1989) J. Exp. Med. 169,
1747–1756
28. Fadeel, B., Thorpe, C. J., Yonehara, S., and Chiodi, F. (1997) Int. Immunol.
9, 201–209
29. Trauth, B. C., Klas, C., Peters, A. M., Matzku, S., Mo¨ller, P., Falk, W.,
Debatin, K. M., and Krammer, P. H. (1989) Science 245, 301–305
30. Dhein, J., Daniel, P. T., Trauth, B. C., Oehm, A., Mo¨ller, P., and Krammer,
P. H. (1992) J. Immunol. 149, 3166–3173
31. Ahrer, K., Buchacher, A., Iberer, G., Josic, D., and Jungbauer, A. (2003)
J. Chromatogr. A 1009, 89–96
32. Siegel, R. M., Frederiksen, J. K., Zacharias, D. A., Chan, F. K., Johnson, M.,
Lynch, D., Tsien, R. Y., and Lenardo, M. J. (2000) Science 288, 2354–2357
Agonist Anti-EDAR Antibodies
SEPTEMBER 2, 2011VOLUME 286• NUMBER 35 JOURNAL OF BIOLOGICAL CHEMISTRY 30779
by guest on January 13, 2016http://www.jbc.org/Downloaded from
Kowalczyk et al. Supplemental Material: Agonist anti-EDAR antibodies Page 1
Online Supplemental Material
Supplemental Figure 1. List of plasmids used in the study.
Amino acids are indicated with the one letter code or, for large stretches of sequence, by
a written description. Signal (signal peptide of haemaglutinin: MAIIYLILLFTAVRG | );
Flag (DYKDDDDK); hEDA1, hEDA2 (SwissProt accession number Q92838); human,
dog, rat, mouse, chicken EDAR (SwissProt accession numbers Q9UNEO, E2RA80,
D3ZGP2, Q9R187 and Q5EFZ7); hFc (IgG1 encoded by GenBank accession number
BC018747 or aa105-330 of SwissProt accession number P01857); hFas (SwissProt
accession number P25445); Ig signal (Signal peptide of mouse Ig, heavy chain:
MNFGFSLIFLVLVLKGVQC | EVKLV). The " | " indicate predicted proteolytic
cleavage sites by signal peptidase. Sequences of interest were cloned into the mammalian
expression vector PCR3 (Invitrogen) or into the retroviral expression vector pMSCV
(Clonetech). Note that ps2765 and ps2783 were obtained by point mutations of the mouse
and human sequences, respectively, and therefore are rat and dog sequences at the amino
acid level only.
Plasmid Designation Protein encoded Vector Figure
ps515 EGFP Enhanced green fluorescent protein PCR3 3
ps1938 Fc-EDA1 Signal-hIgG1 (aa 245-470)-hEDA1 (aa 238-391) PCR3 3, 5, 6
ps1377
pMSCV-puro
Modified pMSCV-puro (Clonetech) with HindIII-BglII-EcoRI-
NotI-XhoI-HpaI-ApaI cloning sites
pMSCV
ps2199 hEDAR:Fas hEDAR (aa 1-183)-VD-hFas (aa 169-335) ps1377 5B
ps2260 mEDAR:Fas mEDAR (aa 1-183)-VD-hFas (aa 169-335) ps1377 5A
ps1235 Fc-EDA2 Signal-LD-hIgG1 (aa 245-470)-hEDA2 (aa 245-389) PCR3 3
ps1431 hEDAR-GPI hEDAR (aa 1-183)-VD-hTRAILR3 (aa 157-259) PCR3 3
ps2783 dogEDAR-GPI dogEDAR (aa 1-183)-VD-hTRAILR3 (aa 157-259) PCR3 3
ps2765 ratEDAR-GPI ratEDAR (aa 1-179)-VD-hTRAILR3 (aa 157-259) PCR3 3
ps1765 mEDAR-GPI mEDAR (aa 1-183)-VD-hTRAILR3 (aa 157-259) PCR3 3
ps2290 chEDAR-GPI chEDAR (aa 1-183)-VD-hTRAILR3 (aa 157-259) PCR3 3
ps548 Flag-EDA1 Signal-Flag-GPGQVQLQVD-mEDA1 (aa 245-391) PCR3 2
ps336 Flag-BAFF Signal-Flag-GPGQVQLQ-hBAFF (aa 137-285) PCR3 2
ps930 hEDAR-Fc hEDAR (aa 1-183)-VD-hIgG1 (aa 245-470) PCR3 2
ps2887 hEDAR (CRD1+2+3)-Fc hEDAR (aa 1-149)-VD-hIgG1 (aa 245-470) PCR3 2
ps2484 hEDAR (CDR1+2)-Fc hEDAR (aa 1-114)-VD-hIgG1 (aa 245-470) PCR3 2
ps2481 hEDAR(CRD1)-Fc hEDAR (aa 1-72)-VD-hIgG1 (aa 245-470) PCR3 2
ps2482 hEDAR (CRD2)-Fc Signal-LE-hEDAR (aa 71-114)-VD-hIgG1 (aa 245-470) PCR3 2
ps2524 hEDAR(CRD3)-Fc Signal-LE-hEDAR (aa 115-149)-VD-hIgG1 (aa 245-470) PCR3 2
ps2515 hEDAR(CRD2+3)-Fc Signal-LE-hEDAR (aa 71-149)-VD-hIgG1 (aa 245-470) PCR3 2
ps2508 hEDAR(CRD3+stalk)-Fc Signal-LE-hEDAR (aa 115-183)-VD-hIgG1 (aa 245-470) PCR3 2
ps2509 hEDAR(stalk)-Fc Signal-LE-hEDAR (aa 150-183)-VD-hIgG1 (aa 245-470) PCR3 2
ps815 mEDAR-Fc mEDAR (aa 1-183)-VD-hIgG1 (aa 245-470) PCR3 2
ps229 hFas-Fc hFas (aa1-170)-VDhIgG1 (aa 245-470) PCR3 2
Kowalczyk et al.
Supplemental Material: Agonist anti-EDAR antibodies Page 2
Supplemental Figure 2: Amino acid sequences of light and heavy chains variable
regions of anti-EDAR monoclonal antibodies.
Sequences start at the mature N-terminus. Complementarity determining regions (CDRs)
are highlighted in boxes. Putative junctions of the protein sections encoded by the V, D
and J genes, or by randomly added nucleotides (N) are indicated. The junction with the
constant region (C) is also shown. Note that the light chains of anti-EDAR antibodies 2
and 4 are identical, and the heavy chains of anti-EDAR antibodies 10 and 11 are
identical. Anti-EDAR antibodies 1, 3 and 8 have similar heavy and light chains, most
probably originating from the same VH and VL genes.
Kowalczyk et al.
Supplemental Material: Agonist anti-EDAR antibodies Page 3
Supplemental Figure 3: Half-life determination of agonistic anti-EDAR antibodies.
Wild type mice were intravenously injected with 200 µl of 1 mg/ml of anti-EDAR 1 or
anti-EDAR 3 antibodies. Serum samples were collected after 20 minutes, 1, 2, 8, 16 and
32 days. The concentration of the anti-EDAR mAb was determined by incubating serial
dilutions of serum in wells coated with human EDAR-Fc at 1 µg/ml, followed by
horseradish peroxidase-coupled anti-mouse IgG and OPD substrate. For analysis, the
serum dilutions giving OD = 1 (considered to represent the EC50) for each time points
were plotted as a function of time. An exponential curve was fitted on the series of points
except the time point 20 minutes. A half-lives of 10 to 11 days were thus determined for
anti-EDAR antibodies 1 and 3.
Kowalczyk et al.
Supplemental Material: Agonist anti-EDAR antibodies Page 4
Supplemental Figure 4: An Fab
2 fragment of an agonist anti-EDAR antibody is active
in vivo, whereas a monomeric Fab fragment acts as an inhibitor.
A. Superdex-200 gel permeation chromatography elution profiles of ficin-generated Fab
and Fab
2 fragments of anti-EDAR antibody 1, previously isolated by size exclusion
chromatography (see Fig. 4A). Main peaks were collected and used for in vivo
experiments.
B. The Fab’2 fragment was injected at 0.5 mg/kg in newborn Tabby mice. Tail hair
formation was recorded 3 weeks later. Administration of the Fab fragment at 4 mg/kg did
not induce tail hair formation (n=3).
C. The Fab’2 fragment was injected ip at 0.5 mg/kg in 3 days-old Tabby pups, which is
one of the latest time point at which tail hair formation can still be partially rescued on
the ventral side of the tail (on the left of the picture). The Fab fragment was injected at 4
mg/kg 4 h before administration of the Fab’2 fragment and at 6 mg/kg together with the
Fab’2 fragment. The Fab was then administered again at 10 mg/kg 24 and 48 h later. Co-
treatment with the Fab fragment prevented the action of the Fab’2 fragment.
Supplemental Figure 5: An anti-EDAR antibody reverts many ectodermal dysplasia
phenotypes in EDA-deficient mice.
A pregnant Tabby mouse was treated iv at day 9 and 17 (E9/E17) of gestation with anti-
EDAR 3 at 16 mg/kg. Offspring was analyzed at 6 months of age, as described in the
legend to Figure 7.
Kowalczyk et al.
Supplemental Material: Agonist anti-EDAR antibodies Page 5
Supplemental Figure 6: Long-term reversion of ectodermal dysplasia symptoms upon
treatment with agonist anti-EDAR antibodies.
A pregnant Tabby mouse was treated iv at day 13 (E13) of gestation with anti-EDAR 3 at
6.5 mg/kg. Offspring and age-matched Tabby mouse were analyzed at 26 months of age.
A. Tail. B. Tail tip, showing absence of a kink in the treated mouse. C. Back hair,
showing the presence of the longer guard hair in the treated mouse. D. Sweat test
showing the presence of functional sweat glands in the treated mouse (arrowhead). E.
Retro-auricular region. F. Eye.
Kowalczyk et al.
Supplemental Material: Agonist anti-EDAR antibodies Page 6
Supplemental Figure 7: Amino acid sequence of the mature extracellular domain of
EDAR in different species.
Divergent amino acids are highlighted with colours. The positions of the cystein-rich
domains (CRDs) and stalk are indicated.
Supplemental Figure 8: Correlation between low dissociation constants and agonist
activity of anti-EDAR antibodies in an in vitro reporter cell line assay.
Binding characteristics of the agonist anti-EDAR monoclonal antibodies (see Table 1)
were plotted against their activity score in vivo (1/EC50, see Table 1) and their activity
score in vitro. The activity score in vitro was determined as 8*(1/log(EC50 hEDAR)) +
5.6*(1/log(EC50 mEDAR)) (see Table 1 for EC50 values). The correction factors 8 and 5.6
were chosen so that the best antibody(ies) in the human or mouse EDAR:Fas assays
score(s) 8 points. Antibodies showing agonist activity in vitro are identified by their
numbers in the graphs.
Stéphane Demotz and Pascal Schneider
Favre, Douglas M. Jefferson, Denis J. Headon,
Giovanna Badic, Anne-Lise Etter, Manuel
Mauldin, Olivier Gaide, Aubry Tardivel,
Willen, Margret L. Casal, Elizabeth A.
Christine Kowalczyk, Nathalie Dunkel, Laure
Antibodies
Receptor (EDAR) Agonist Monoclonal
Characterization of Anti-ectodysplasin A
Molecular and Therapeutic
Developmental Biology:
doi: 10.1074/jbc.M111.267997 originally published online July 5, 2011
2011, 286:30769-30779.J. Biol. Chem.
10.1074/jbc.M111.267997Access the most updated version of this article at doi:
.JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
Supplemental material:
http://www.jbc.org/content/suppl/2011/07/05/M111.267997.DC1.html
http://www.jbc.org/content/286/35/30769.full.html#ref-list-1
This article cites 32 references, 13 of which can be accessed free at
by guest on January 13, 2016http://www.jbc.org/Downloaded from

Supplementary resources (29)

... Indeed, three human male fetuses carrying EDA null mutations who received Fc-EDA via the intra-amniotic route benefitted from restoration of their sweating ability, with beneficial outcome also on teeth and salivary glands [9]. Antibodies that target the EDA-EDAR pathway in positive or negative ways can prevent or induce ectodermal dysplasia phenotypes in animal models [11,12]. In Eda-deficient mice, in utero treatment of affected fetuses with an anti-EDAR antibody via the transplacental route restored nasal submucosal glands and the paired nasopharyngeal submucosal glands that protect the opening of each of the Eustachian tubes. ...
... 2. Agonist anti-EDAR mouse monoclonal IgG1 antibodies (mAbEDAR1 or mAbEDAR3) or function-blocking anti-EDA mouse monoclonal IgG1 antibodies (EctoD2 or EctoD3): Purify them from conditioned media of hybridoma cultures by affinity chromatography on Protein G (GE Healthcare) [11,12]. Store the antibodies frozen at À70 C in sterile PBS at concentrations of 1 to 5 mg/mL until use. ...
... The thread should look like a spiral ( Fig. 12.4a, g). 11. For the last stitch, pull the needle until there is a loop of about 2 cm of thread remaining on the right side of the wound (Fig. 12.4h). ...
Chapter
Genetic deficiency of ectodysplasin A (EDA) causes X-linked hypohidrotic ectodermal dysplasia, a congenital condition characterized by the absence or abnormal formation of sweat glands, teeth, and several skin appendages. Stimulation of the EDA receptor (EDAR) with agonists in the form of recombinant EDA or anti-EDAR antibodies can compensate for the absence of Eda in a mouse model of Eda deficiency, provided that agonists are administered in a timely manner during fetal development. Here we provide detailed protocols for the administration of EDAR agonists or antagonists, or other proteins, by the intravenous, intraperitoneal, and intra-amniotic routes as well as protocols to collect blood, to visualize sweat gland function, and to prepare skulls in mice.
... Our investigations and related studies clearly validate the hypothesis that de novo repression of target genes, such as Usag-1, could be used to stimulate arrested tooth germs in order to induce new tooth formation in mammals. Indeed, in animal models of EDA deficiency, which is associated with the human disorder hypohidrotic ectodermal dysplasia which involves hypodontia, the administration of a soluble EDA receptor agonist has been shown to correct many phenotypic abnormalities, including abnormalities of the dentition in mice (primary) and dogs (secondary or permanent) [44][45][46][47]. ...
... After a single postnatal systemic administration of recombinant EDA or anti-EDA receptor agonist monoclonal antibody, it was confirmed that all the physiological aspects of missing teeth, such as the number, size, shape, location, and timing of eruption, were restored, and the teeth functioned normally [44,47]. In fact, lifelong phenotypic correction was achieved after a short course of treatment [44,47]. ...
... After a single postnatal systemic administration of recombinant EDA or anti-EDA receptor agonist monoclonal antibody, it was confirmed that all the physiological aspects of missing teeth, such as the number, size, shape, location, and timing of eruption, were restored, and the teeth functioned normally [44,47]. In fact, lifelong phenotypic correction was achieved after a short course of treatment [44,47]. In the future, molecular targeted therapies could be used to generate teeth in patients with congenital tooth agenesis by stimulating arrested tooth germs (Fig. 1). ...
Article
Full-text available
Analysis of various genetically modified mice, with supernumerary teeth, has revealed the following two intrinsic molecular mechanisms that increase the number of teeth. One plausible explanation for supernumerary tooth formation is the rescue of tooth rudiments. Topical application of candidate molecules could lead to whole tooth formation under suitable conditions. Congenital tooth agenesis is caused by the cessation of tooth development due to the deletion of the causative gene and suppression of its function. The arrest of tooth development in Runx2 knockout mice, a mouse model of congenital tooth agenesis, is rescued in double knockout mice of Runx2 and Usag-1. The Usag-1 knockout mouse is a supernumerary model mouse. Targeted molecular therapy could be used to generate teeth in patients with congenital tooth agenesis by stimulating arrested tooth germs. The third dentition begins to develop when the second successional lamina is formed from the developing permanent tooth in humans and usually regresses apoptotically. Targeted molecular therapy, therefore, seems to be a suitable approach in whole-tooth regeneration by the stimulation of the third dentition. A second mechanism of supernumerary teeth formation involves the contribution of odontogenic epithelial stem cells in adults. Cebpb has been shown to be involved in maintaining the stemness of odontogenic epithelial stem cells and suppressing epithelial-mesenchymal transition. Odontogenic epithelial stem cells are differentiated from one of the tissue stem cells, enamel epithelial stem cells, and odontogenic mesenchymal cells are formed from odontogenic epithelial cells by epithelial-mesenchymal transition. Both odontogenic epithelial cells and odontogenic mesenchymal cells required to form teeth from enamel epithelial stem cells were directly induced to form excess teeth in adults. An approach for the development of targeted therapeutics has been the local application of monoclonal neutralizing antibody/siRNA with cationic gelatin for USAG-1 or small molecule for Cebpb.
... XLHED is a systemic genetic disease caused by mutation of the EDA gene and deficiency of the signaling protein EDA [79,80], which leads to the abnormal development of exocrine glands, hair and teeth [81]. The affected individuals inescapably suffer from severe MGD and a dry eye phenotype [53], along with chronic conjunctivitis and blepharitis [33]. ...
... Additionally, after the Fc: EDA1 treatment during the postnatal period, meibomian gland and eyelid development were successful and tear production was significantly increased. Meanwhile, the rate of keratoconjunctivitis sicca and eye infection incidence was greatly reduced [63,64,80]. Taken together, these results indicated that the soluble recombinant EDA (Fc: EDA1) applied during the postnatal period can efficiently maintain the homeostasis of the ocular surface. ...
Article
Full-text available
Ectodysplasin A (EDA), a ligand of the TNF family, plays an important role in maintaining the homeostasis of the ocular surface. EDA is necessary for the development of the meibomian gland, the lacrimal gland, as well as the proliferation and barrier function of the corneal epithelium. The mutation of EDA can induce the destruction of the ocular surface resulting in keratopathy, abnormality of the meibomian gland and maturation of the lacrimal gland. Experimental animal studies showed that a prenatal ultrasound-guided intra-amniotic injection or postnatal intravenous administration of soluble recombinant EDA protein can efficiently prevent the development of ocular surface abnormalities in EDA mutant animals. Furthermore, local application of EDA could restore the damaged ocular surface to some extent. Hence, a recombinant EDA-based therapy may serve as a novel paradigm to treat ocular surface disorders, such as meibomian gland dysfunction and corneal epithelium abnormalities.
... The Tabby (Eda Ta ) mouse is a model of X-linked HED (XLHED) and is deficient in EDA. Treatment of adult Eda Ta mice with agonist anti-EDAR antibody (Kowalczyk et al., 2011; restores sebaceous gland growth and sebum production, and sustained treatment of Eda Ta and wildtype mice heightens sebum production (Kowalczyk-Quintas et al., 2015). In addition, prenatal correction of XLHED with a recombinant protein that includes the receptor binding domain of EDA restores Meibomian gland growth in humans (Schneider et al., 2018) and dogs (Margolis et al., 2019). ...
... Eda Ta mice were administered agonist anti-EDAR antibody (mAbEDAR1) (Kowalczyk et al., 2011;Schuepbach-Mallepell et al., 2021) at 2 mg/kg either prenatally (E10.5 and E17.5) (n=2) or prenatally and postnatally (E10.5, E17.5, P1, P7 and P14; n=6), and phenotyped at P21. In addition, Eda Ta mice treated prenatally (E10.5 and E17.5) with anti-EDAR antibody (n=9) or with isotype control antibody (Aprily2) (n=10) were phenotyped at P85 or P82-P84, respectively (see details of administration routes in del-Pozo et al., 2019a). ...
Article
Full-text available
In mice, rats, dogs and humans the growth and function of sebaceous glands and eyelid Meibomian glands depend on the ectodysplasin signalling pathway. Mutation of genes encoding the ligand EDA, its transmembrane receptor EDAR, and the intracellular signal transducer EDARADD leads to Hypohidrotic Ectodermal Dysplasia characterised by impaired development of teeth and hair as well as cutaneous glands. The rodent ear canal has a large auditory sebaceous gland, the Zymbal's gland, whose function in the health of the ear canal and tympanic membrane has not been determined. We report that the EDA deficient Tabby (EdaTa) mouse, the EDAR deficient mouse (EdarOVE1B/OVE1B) and the EDARADD deficient sparse and wavy hair rat (Edaraddswh/swh) have Zymbal's gland hypoplasia. EdaTa mice also have ear canal hypotrichosis and a 25% prevalence of otitis externa at P21. Treatment with agonist anti-EDAR antibodies rescues Zymbal's glands and ear canal pilosebaceous units. The aetiopathogenesis of otitis externa involves infection with Gram-positive cocci and dosing pregnant and lactating EdaTa females and pups with Enrofloxacin reduces the prevalence of otitis externa. We infer the deficit of sebum is the principal factor in predisposition to bacterial infection and the EdaTa mouse is a potentially useful microbial challenge model for human acute otitis externa, commonly known as swimmer's ear.
... Several previous studies have shown that high EDAR expression promotes more hair follicles in EDAR-hyperactivated mice [33] and geneedited Cashmere goats [37]. In addition, hair formation of EDAdeficient mice was induced by administering anti-EDAR Agonist Monoclonal Antibodies, which revealed a strong association between EDAR and hair follicle density [38]. EDAR370A (1540T/C, 370Val/Ala) knock-in mice showed reduced EDAR expression and exhibited a larger proportion of thick hairs compared to wildtype mice [39,40]. ...
Article
Full-text available
Introduction: Fine-wool sheep are the most common breed used by the wool industry worldwide. Fine-wool sheep have over a three-fold higher follicle density and a 50% smaller fiber diameter than coarse-wool sheep. Objectives: This study aims to clarify the underlying genetic basis for the denser and finer wool phenotype in fine-wool breeds. Method: Whole-genome sequences of 140 samples, Ovine HD630K SNP array data of 385 samples, including fine, semi-fine, and coarse wool sheep, as well as skin transcriptomes of nine samples were integrated for genomic selection signature analysis. Results: Two loci at keratin 74 (KRT74) and ectodysplasin receptor (EDAR) were revealed. Fine-scale analysis in 250 fine/semi-fine and 198 coarse wool sheep narrowed this association to one C/A missense variant of KRT74 (OAR3:133,486,008, P = 1.02E-67) and one T/C SNP in the regulatory region upstream of EDAR (OAR3:61,927,840, P = 2.50E-43). Cellular over-expression and ovine skin section staining assays confirmed that C-KRT74 activated the KRT74 protein and specifically enlarged cell size at the Huxley's layer of the inner root sheath (P < 0.01). This structure enhancement shapes the growing hair shaft into the finer wool than the wild type. Luciferase assays validated that the C-to-T mutation upregulated EDAR mRNA expression via a newly created SOX2 binding site and potentially led to the formation of more hair placodes. Conclusions: Two functional mutations driving finer and denser wool production were characterized and offered new targets for genetic breeding during wool sheep selection. This study not only provides a theoretical basis for future selection of fine wool sheep breeds but also contributes to improving the value of wool commodities.
... For the inhibition of ectodysplasin signaling, stage Greenbaum 15 (G15, Greenbaum 2002) and G16 embryo explants were cultured for 5 days with 2 μg/ml EctoD2, a blocking anti-EDA mouse monoclonal antibody previously shown to phenocopy complete Eda deficiency in mice, and to be successful in inhibiting feather bud formation in chicken skin (Ho et al., 2019;Kowalczyk-Quintas et al., 2014). Embryo explants were also cultured under identical conditions with an agonist anti-Edar antibody (mAbEdar1) able to rescue Eda-deficiency in mice (Kowalczyk et al., 2011), or with an isotype-matched control antibody (Aprily2). To examine the effect of Activin A on turtle scute development, stage G15 and G16 embryo explants were cultured for 5 days with 500 ng/ml activin A protein (courtesy of Marko Hyvönen; Harrington et al., 2006) ...
Article
The scutes of the turtle shell are epidermal shields that begin their formation during the early stages of shell development. Like other skin appendages, turtle scutes are hypothesized to be patterned by reaction-diffusion systems. We have previously established ex vivo and in silico systems to study these mechanisms experimentally and have further shown that mathematical models can explain the dynamics of the induction of turtle scute primordia and the generation of final scute architecture. Using these foundations, we expand our current knowledge and test the roles of ectodysplasin and activin signaling in the development of turtle scutes. We find that these molecules play important roles in the prepatterning of scute primordia along the carapacial ridge and show that blocking Edar signaling may lead to a complete loss of marginal scute primordia. We show that it is possible to reproduce these observations using simple mathematical modeling, thereby suggesting a stabilizing role for ectodysplasin within the reaction-diffusion mechanisms. Finally, we argue that our findings further entrench turtle scutes within a class of developmental systems composed of hierarchically nested reaction-diffusion mechanisms, which is conserved across ectodermal organs.
... These alleles may increase the incidence or progression of breast tumours with squamous metaplasia in these populations. A number of recombinant protein and antibody reagents to stimulate and repress the EDAR pathway in vivo have been developed recently [44,45], with one of the stimulators in clinical trial for early-life correction of HED caused by EDA loss of function [46]. Such agents may represent useful modalities for future diagnosis and treatment of breast cancer with squamous metaplasia. ...
Article
Full-text available
Ectodysplasin A receptor (EDAR) is a death receptor in the Tumour Necrosis Factor Receptor (TNFR) superfamily with roles in the development of hair follicles, teeth and cutaneous glands. Here we report that human Oestrogen Receptor (ER) negative breast carcinomas which display squamous differentiation express EDAR strongly. Using a mouse model with a high Edar copy number, we show that elevated EDAR signalling results in a high incidence of mammary tumours in breeding female mice. These tumours resemble the EDAR -high human tumours in that they are characterised by a lack of oestrogen receptor expression, contain extensive squamous metaplasia, and display strong β-catenin transcriptional activity. In the mouse model, all of the tumours carry somatic deletions of the third exon of the CTNNB1 gene that encodes β-catenin. Deletion of this exon yields unconstrained β-catenin signalling activity. We also demonstrate that β-catenin activity is required for transformed cell growth, showing that increased EDAR signalling creates an environment in which β-catenin activity can readily promote tumourigenesis. Together, this work identifies a novel death receptor oncogene in breast cancer, whose mechanism of transformation is based on the interaction between the WNT and Ectodysplasin A (EDA) pathways.
Article
Surface plasmon resonance (SPR) is an optical technique that is utilized for detecting molecular interactions that occur in direct protein-protein interactions. Binding of a mobile molecule (analyte) to a molecule immobilized on a thin metal film (ligand) changes the refractive index of the film. The angle of extinction of light that is completely reflected, after polarized light impinges upon the surface, is altered and monitored as a change in detector position for a dip in reflected intensity (the surface plasmon resonance phenomenon). Because the method strictly detects mass, there is no need to label the interacting components, thus eliminating possible changes of their molecular properties. One of the advantages in SPR is its high sensitivity, compatible with the need for purification of small amounts of protein for analysis. This chapter concentrates on practical methodologies for performing surface plasmon resonance analysis.
Thesis
Hintergründe und Ziel der Arbeit Das Signalprotein Ektodysplasin A (EDA) gehört zu den Tumornekrosefaktor-Liganden und existiert in zwei unterschiedlichen Spleißvarianten (EDA1 und EDA2). EDA1 bindet an den auf Chromosom 2 kodierten EDA-Rezeptor (EDAR), EDA2 an den X-chromosomalen EDA-Rezeptor (XEDAR/EDA2R). Mutationen, die zur Inaktivierung von EDA1 oder EDAR führen, lösen eine hypohidrotische ektodermale Dysplasie (HED) aus, die durch die Fehlanlage von Zähnen, Haaren und Haut- sowie Schleimhautdrüsen gekennzeichnet ist. EDA1 spielt bereits während der embryonalen Plakodenstadien eine wichtige Rolle bei der Entwicklung vom Ektoderm abstammender Strukturen und ist wahrscheinlich auch im Erwachsenenalter noch für die Funktion mehrerer Hautanhangsgebilde von Bedeutung. Da die HED besonders wegen des Hyperthermierisikos eine lebensbedrohliche Krankheitsgruppe darstellt, besteht großes Interesse, sie so früh wie möglich zu diagnostizieren. Ziel dieser Arbeit war es, eine Methode zur Bestimmung von EDA in Nabelschnurblut- und Speichelproben zu etablieren und somit den Weg zur Frühdiagnose der häufigsten Form der HED zu ebnen. Methoden Zur Verfügung standen insgesamt 42 Speichel- und Serumproben von an X-chromosomaler HED (XLHED) erkrankten Erwachsenen, heterozygoten Trägern einer EDA-Mutation und gesunden Probanden. Des Weiteren wurde Nabelschnurblut von 12 Neugeborenen und drei Frühgeborenen sowie Serum von 6 Mäusen mit unterschiedlichen Mutationen (EDA-defizient, EDAR-Nullmutation, EDA-transgene Mäuse) untersucht. Mit Hilfe verschiedener monoklonaler Antikörper (Anti-EDA-Antikörper EctoD2, EctoD3 und Renzo-2) sowie rekombinanter Proteine (EDAR-Fc, XEDAR-Fc, BMCA-Fc und Fc-EDA1) konnte EDA mittels Western Blot und AlphaLISA detektiert und quantifiziert werden. Ergebnisse In Nabelschnurblut wurden drei- bis viermal höhere EDA-Konzentrationen gemessen als in Serumproben von Erwachsenen. Den größten messbaren Anteil repräsentierte rezeptorbindungsfähiges EDA1, jedoch konnte auch eine kleinere Fraktion von EDA2 detektiert werden. Seren EDA-defizienter Patienten wiesen entweder nur hintergrundähnliche oder sehr geringe Konzentrationen des Signalproteins auf, welches nicht in der Lage war, an den rekombinanten EDAR zu binden. Serumproben eines Patienten mit der Missense-Mutation V262F, die eine milde Form der XLHED bewirkt, enthielten geringe Mengen an EDA, welches aber mit EDAR normal interagierte. Bei zwei von der Krankheit nur leicht betroffenen heterozygoten Trägerinnen einer EDA-Mutation wurden intermediäre EDA-Konzentrationen detektiert, während eine schwer betroffene Mutationsträgerin kein EDA aufwies. Auch im Speichel gesunder Erwachsener ließen sich kleine Mengen an EDA nachweisen. Schließlich gelang es, in auf Filterpapier getrocknetem Blut von gesunden Erwachsenen und Neugeborenen die EDA-Konzentration zu bestimmen, die jeweils deutlich höher war als im Blut von EDA-defizienten Patienten. Schlussfolgerung Die hier beschriebene Methode zur Detektion von EDA, kombiniert mit einem Rezeptorbindungs-Assay, könnte die Frühdiagnostik kompletter oder partieller EDA-Defizienz anhand eines getrockneten Bluttropfens ermöglichen und so für die adäquate Versorgung von Säuglingen mit XLHED sehr hilfreich sein.
Article
Full-text available
We have prepared an mAb specific for a human cell surface component (termed anti-Fas mAb). Anti-Fas shows cell-killing activity that is indistinguishable from the cytolytic activity of TNF. Fas antigen was characterized by western blotting, indicating that Fas antigen is a cell surface protein with a molecular weight of 200,000, which is different from the molecular weight of TNF-R. Fas antigen, however, is co-downregulated with the TNF-R when cells sensitive to the cytolytic activity of TNF are incubated with either TNF or anti-Fas. In contrast, Fas antigen on cells insensitive to TNF is not co-downregulated with the TNF-R. We suggest that the cell-killing activity of TNF is mediated by Fas antigen associated with the TNF-R.
Article
Full-text available
Heterozygous mutations encoding abnormal forms of the death receptor Fas dominantly interfere with Fas-induced lymphocyte apoptosis in human autoimmune lymphoproliferative syndrome. This effect, rather than depending on ligand-induced receptor oligomerization, was found to stem from ligand- independent interaction of wild-type and mutant Fas receptors through a specific region in the extracellular domain. Preassociated Fas complexes were found in living cells by means of fluorescence resonance energy transfer between variants of green fluorescent protein. These results show that formation of preassociated receptor complexes is necessary for Fas signaling and dominant interference in human disease.
Article
Full-text available
Mutations in the TNF family ligand EDA1 cause X-linked hypohidrotic ectodermal dysplasia (XLHED), a condition characterized by defective development of skin appendages. The EDA1 protein displays a proteolytic processing site responsible for its conversion to a soluble form, a collagen domain, and a trimeric TNF homology domain (THD) that binds the receptor EDAR. In-frame deletions in the collagen domain reduced the thermal stability of EDA1. Removal of the collagen domain decreased its activity about 100-fold, as measured with natural and engineered EDA1-responsive cell lines. The collagen domain could be functionally replaced by multimerization domains or by cross-linking antibodies, suggesting that it functions as an oligomerization unit. Surprisingly, mature soluble EDA1 containing the collagen domain was poorly active when administered in newborn, EDA-deficient (Tabby) mice. This was due to a short stretch of basic amino acids located at the N terminus of the collagen domain that confers EDA1 with proteoglycan binding ability. In contrast to wild-type EDA1, EDA1 with mutations in this basic sequence was a potent inducer of tail hair development in vivo. Thus, the collagen domain activates EDA1 by multimerization, whereas the proteoglycan-binding domain may restrict the distribution of endogeneous EDA1 in vivo.
Article
Full-text available
Mutations of the Ectodysplasin-A (EDA) gene are generally associated with the syndrome hypohidrotic ectodermal dysplasia (MIM 305100), but they can also manifest as selective, non-syndromic tooth agenesis (MIM300606). We have performed an in vitro functional analysis of six selective tooth agenesis-causing EDA mutations (one novel and five known) that are located in the C-terminal tumor necrosis factor homology domain of the protein. Our study reveals that expression, receptor binding or signaling capability of the mutant EDA1 proteins is only impaired in contrast to syndrome-causing mutations, which we have previously shown to abolish EDA1 expression, receptor binding or signaling. Our results support a model in which the development of the human dentition, especially of anterior teeth, requires the highest level of EDA-receptor signaling, whereas other ectodermal appendages, including posterior teeth, have less stringent requirements and form normally in response to EDA mutations with reduced activity.
Article
Full-text available
We have prepared an mAb specific for a human cell surface component (termed anti-Fas mAb). Anti-Fas shows cell-killing activity that is indistinguishable from the cytolytic activity of TNF. Fas antigen was characterized by western blotting, indicating that Fas antigen is a cell surface protein with a molecular weight of 200,000, which is different from the molecular weight of TNF-R. Fas antigen, however, is co-downregulated with the TNF-R when cells sensitive to the cytolytic activity of TNF are incubated with either TNF or anti-Fas. In contrast, Fas antigen on cells insensitive to TNF is not co-downregulated with the TNF-R. We suggest that the cell-killing activity of TNF is mediated by Fas antigen associated with the TNF-R.
Article
Mouse Tabby (Ta) and X chromosome-linked human EDA share the features of hypoplastic hair, teeth, and eccrine sweat glands. We have cloned the Ta gene and find it to be homologous to the EDA gene. The gene is altered in two Ta alleles with a point mutation or a deletion. The gene is expressed in developing teeth and epidermis; no expression is seen in corresponding tissues from Ta mice. Ta and EDA genes both encode alternatively spliced forms; novel exons now extend the 3′ end of the EDA gene. All transcripts recovered have the same 5′ exon. The longest Ta cDNA encodes a 391-residue transmembrane protein, ectodysplasin-A, containing 19 Gly-Xaa-Yaa repeats. The isoforms of ectodysplasin-A may correlate with differential roles during embryonic development.
Article
Ectodermal dysplasias are a group of congenital disorders with defective development of the epidermis and its appendages. X-linked hypohydrotic ectodermal dysplasia (XLHED; OMIM 305100) is the most common form of ectodermal dysplasia. We report on two monozygotic twin girls with XLHED due to a t(X;9) translocation causing a disruption of the EDA gene and non random inactivation of the normal X chromosome. One of the girls died unexpectedly at 2.5 years of age. Autopsy revealed that lack of normal tracheobronchial secretions leading to complete tracheal obstruction by mucous debris was the probable cause of death. Conclusion Morbidity and mortality of ectodermal dysplasias in infancy and early childhood can be significant. Early diagnosis by paediatricians is important and complications should be anticipated.
Article
Patients with defective ectodysplasin A (EDA) have X-linked hypohidrotic ectodermal dysplasia (XLHED; OMIM#305100), a condition comprising hypotrichosis, inability to sweat, abnormal teeth, and frequent pulmonary infections. The XLHED dogs show the same clinical signs as humans with the disorder, including frequent respiratory infections that can be fatal. The respiratory disease in humans and dogs is thought to be due to the absence of tracheal and bronchial glands which are a vital part of the mucociliary clearance mechanism. In our XLHED model, the genetically missing EDA was replaced by postnatal intravenous administration of recombinant EDA resulting in long-term, durable corrective effect on adult, permanent dentition. After treatment with EDA, significant correction of the missing tracheal and bronchial glands was achieved in those dogs that received higher doses of EDA. Moreover, successful treatment resulted in the presence of esophageal glands, improved mucociliary clearance, and the absence of respiratory infection. These results demonstrate that a short-term treatment at a neonatal age with a recombinant protein can reverse a developmental disease and result in vastly improved quality of life.
Article
A total of 27 women of a Brazilian kindred are described as having one or more signs of the Christ-Siemens-Touraine syndrome. The history and physical examination were supplemented by four sweat tests and dermatolglyphic analysis. It is suggested that this syndrome has two forms -- a major form (in males) and a minor one (in females). Two signs verified in some of our patients (mosaic patchy distribution of body hair and radial deviation of distal phalanges of index fingers) seem to be here described for the first time. A review of the literature shows a corrected sex ratio between 1 M: 1.21 F and 1 M: 2.38 F among affecteds. Since the manifestation rate of the gene among carriers was estimated at about 0.70, the actual sex ratio is expected to be not lower than 1 M: 1.40 F. Contrary to a general opinion, affected females outnumber affected males.
Article
Apoptosis, programmed cell death, was previously shown to be induced by the mAb anti-APO-1 (IgG3, kappa) by binding to the APO-1 cell surface Ag, a new member of the nerve growth factor/TNF receptor superfamily. To investigate the role of the Ig H chain Fc regions we compared induction of apoptosis by the original mAb IgG3 anti-APO-1 with anti-APO-1 F(ab')2 fragments and different anti-APO-1 isotypes (IgG1, IgG2b, IgG2a, and IgA) isolated by sequential sublining. We found that IgG3 was the most active isotype; IgG1, IgG2a, and IgA showed intermediate activity, and IgG2b and F(ab')2 were inactive. Cytotoxic activity of the inactive or less active antibody preparations was fully reconstituted by protein A, anti-mouse Ig, or anti-mouse Ig F(ab')2, respectively. Thus, APO-1-mediated induction of apoptosis was dependent on efficient cross-linking of APO-1 cell surface Ag, indirectly augmented by anti-APO-1 Fc-Fc self-aggregation. Because of their different in vitro activity we selected IgG3-, IgG2b-, and IgA anti-APO-1 to test their antitumor activity against solid human B lymphoblastoid tumors in SCID mice. The isotypes showed a different serum half-life (IgG3: 9.2-10.4 days, IgG2b: 1.9-2.6 days, and IgA: 14.1-29.2 h) and a different initial tumor localization 4 h after i.p. injection (IgG3 around the blood vessels, IgG2b homogeneously, and IgA heterogeneously distributed in the tumor). All antibody preparations induced tumor regression by induction of apoptosis, even IgG2b anti-APO-1 inactive in vitro without cross-linking. The activity of IgA anti-APO-1, which did not mediate complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity indicates that apoptosis may be used as the main if not the only mechanism of induction of tumor regression in vivo. As with in vitro, IgG3 anti-APO-1 was the most effective isotype also in vivo. This result suggests that cross-linking of APO-1 on the tumor cell surface may also be required for tumor regression by apoptosis in vivo. Taken together, our data show that selective targeting of apoptosis to tumors may be an efficient antitumor mechanism.