Tải bản đầy đủ (.pdf) (12 trang)

Tài liệu Báo cáo khoa học: Upregulation of the a-secretase ADAM10 – risk or reason for hope? docx

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (290.64 KB, 12 trang )

REVIEW ARTICLE
Upregulation of the a-secretase ADAM10 – risk or reason
for hope?
Kristina Endres and Falk Fahrenholz
Department of Psychiatry and Psychotherapy, Clinical Research Group, Johannes Gutenberg-University, Mainz, Germany
Identification of ADAM10 as a
functional a-secretase
A disintegrin and metalloproteinase 10 (ADAM10)
originally came into focus in genetical and biochemical
research as a peptide sequence purified from bovine
brain myelin membrane preparations [1], and was
referred to as MADM (i.e. mammalian disintegrin-me-
talloprotease). Accidentally, this metalloproteinase was
identified via an artifact resulting from in vitro studies:
it has been described as a proteinase for the cytosolic
myelin basic protein [2], which is a rather unphysiolog-
ical substrate for the type I transmembrane enzyme
ADAM10. Further studies revealed that ADAM10 is
expressed in a wide variety of tissues either in Bos
taurus [3] and, more interestingly, in distinct areas of
the human brain [4,5] and peripheral structures [6,7].
Striking similarity concerning the inhibitory profile of
ADAM10 [8] with the putative a-secretase [9] sug-
gested a more physiological role for its enzymatic
activity: overexpression of the ADAM10 cDNA in
HEK293 cells first identified its function as an amyloid
precursor protein (APP) cleaving a-secretase [8], which
subsequently was verified in vivo. Alzheimer’s disease
(AD) model mice, which were crossbred with
ADAM10 transgenic mice, revealed a strongly attenu-
ated plaque pathology and an enhanced production


of the a-secretase derived soluble cleavage product
APPs-a [10]. Furthermore, these mice had an increased
learning and memory potential [10], which might
correlate with the observed enhanced cholinergic and
Keywords
alpha-secretase; amyloid precursor protein;
Alzheimer’s disease; domain structure;
neuroprotection; shedding; synaptogenesis;
TACE
Correspondence
K. Endres and F. Fahrenholz, Department of
Psychiatry and Psychotherapy, Clinical
Research Group, Johannes Gutenberg-
University, 55131 Mainz, Germany
Fax: + 49 6131 176690
Tel: + 49 6131 172133
E-mail:
uni-mainz.de;
(Received 4 November 2009, revised 10
December 2009, accepted 6 January 2010)
doi:10.1111/j.1742-4658.2010.07566.x
A decade ago, a disintegrin and metalloproteinase 10 (ADAM10) was iden-
tified as an a-secretase and as a key proteinase in the processing of the amy-
loid precursor protein. Accordingly, the important role that it plays in
Alzheimer’s disease was manifested. Animal models with an overexpression
of ADAM10 revealed a beneficial profile of the metalloproteinase with
respect to learning and memory, plaque load and synaptogenesis. Therefore,
ADAM10 presents a worthwhile target with respect to the treatment of a
neurodegenerative disease such as Morbus Alzheimer. Initially, ADAM10
was suggested to be an enzyme, shaping the extracellular matrix by cleavage

of collagen type IV, or to be a tumour necrosis factor a convertase. In a rel-
atively short time, a wide variety of additional substrates (with amyloid pre-
cursor protein probably being the most prominent) has been identified and
the search is still ongoing. Hence, any side effects concerning the therapeutic
enhancement of ADAM10 a-secretase activity have to be considered. The
present review summarizes our knowledge about the structure and function
of ADAM10 and highlights the opportunities for enhancing the expression
and ⁄ or activity of the a-secretase as a therapeutic target.
Abbreviations
5-HT4, serotonin 5-hydroxytryptamine; AD, Alzheimer’s disease; ADAM, a disintegrin and metalloproteinase; APP, amyloid precursor protein;
Ab, b-amyloid protein; GPCR, G protein-coupled receptor; GPI, glycosylphosphatidylinositol; PACAP, pituitary adenylate cyclase-activating
peptide; PKC, protein kinase C; SH3, Src homology 3; TACE, tumour necrosis factor a cleaving enzyme.
FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS 1585
glutamatergic synaptogenesis [11]. By contrast, mice
with a dominant negative mutant of ADAM10 had
lowered amounts of APPs-a, accompanied by an
enhanced amount of plaques [10] and learning deficien-
cies in the Morris water maze test [12]. In summary,
what began with a fallacious observation ended up
with the discovery of an enzyme that might have impli-
cations for a therapeutic approach in AD [13–15].
Protein structure and gene
organization of ADAM10
The enzyme ADAM10 belongs to the subgroup of
metzincins within the zinc proteinases family. The typi-
cal multidomain structure of ADAM10 as a type I
integral transmembrane protein consists of a prodo-
main, a catalytical domain with a conserved zinc bind-
ing sequence, a cysteine-rich disintegrin-like domain, a
transmembrane domain and a rather short cytoplasmic

domain (Fig. 1).
The nascent protein itself is not functional and is
produced as a zymogene. After cleavage of the signal-
ling sequence, ADAM10 enters the secretory pathway
to be processed and thereby activated by the propro-
tein convertases furin or PC7 [16]. This constitutive
processing has been demonstrated for the prodomains
of several ADAMs [17–19]. Regarding ADAM10, the
prodomain was revealed to exhibit a dual function: the
separately expressed prodomain was capable of inacti-
vating endogenous ADAM10 in cell culture experi-
ments but overexpressed ADAM10 without its
prodomain was inactive [16]. By contrast, coexpression
of the prodomain in trans rescued the activity of the
deletion mutant of ADAM10 without the intramolecu-
lar prodomain [16]. In addition, the recombinant mur-
ine prodomain purified from Escherichia coli acts as a
potent and selective competitive inhibitor in experi-
ments performed in vitro [20]. This implicates that the
prodomain of ADAM10 acts not only as a transient
inhibitor, but also as an internal chaperone in the mat-
uration of the enzyme. Accordingly, the viral delivery
of furin into the brain of AD model mice increased
a-secretase activity and reduced b-amyloid protein
(Ab) production in infected brain regions [21], demon-
strating the in vivo relevance of the removal of the
prodomain of ADAM10. Recently, by reciprocal coim-
munoprecipitation, tetraspanin 12 was identified as an
interaction partner for ADAM10 that enhances a-sec-
retase shedding of APP, probably by regulating matu-

ration of the prodomain of ADAM10 [22].
The catalytical domain of ADAM10 contains a
typical zinc-binding consensus motif (HEXGHXX
GXXHD; Fig. 1) and the point mutation E384A,
which compromises this motif, leads to a substantial
decrease in APPs-a secretion in HEK cells and in mice
[10,23]. Glycosylation sites in the catalytic and disinte-
grin domain contain high-mannose as well as complex-
type N-glycans, and a mutation at the N-glycosylation
site N439 increased ADAM10s susceptibility to proteo-
lytical degradation [24].
Although the removal of the disintegrin domain of
ADAM10 did not grossly affect shedding of APP in
cell culture experiments [23], cleavage of some sub-
strate molecules is likely to be influenced by noncata-
lytical domains. For example, epidermal growth factor
3
708
|
PKLPPPKPLPGTLKRRRPPQPIQQPQRQRPR

pat.7


pat.4

bipartite
1
2
4

5
210
|
RKKR
383
|
HEVGHNFGSPHD
Fig. 1. Domain structure of human ADAM10. ADAM10 is com-
posed of five different domains: the prodomain (1) has bifunctional
properties as an intramolecular chaperone and as an inhibitor of the
catalytic function in the zymogene. By detaching the prodomain via
proprotein convertase cleavage (recognition motif shown), the cata-
lytic domain with the conserved zinc binding motiv (2) becomes
activated. A mutation of the glutamate residue at position 384
(highlighted) into an alanine leads to a dominant-negative mutant of
the enzyme. The cystein-rich disintegrin domain (3) is followed by a
transmembrane region (4). In the intracellular space, a short cyto-
plasmic domain protrudes (5), which contains important sequence
motives for protein localization (SH3 motifs highlighted) [28,29]. In
addition, nuclear localization sequences have been assumed
because the ADAM10 intracellular domain was found to translocate
to the nucleus [41,79]: PSORTII analysis indicates two pattern 4,
one pattern 7 and one bipartite nuclear localization sequence
(underlined).
Upregulation of ADAM10 K. Endres and F. Fahrenholz
1586 FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS
cleavage is at least partially impaired in ADAM10
) ⁄ )
cells overexpressing a cytoplasmic domain deletion
mutant of ADAM10 [25]. In accordance with this find-

ing, the cytoplasmic domain of ADAM10 contains an
IQ consensus binding site for calmodulin that afflicts
maturation of the proteinase [25]. Additionally,
ADAM10 has been shown to be activated by a
calcium ionophore and the calmodulin inhibitor triflu-
oroperazine [26,27]. The cytoplasmic domain of
ADAM10 furthermore contains two proline-rich puta-
tive Src homology 3 (SH3) binding domains, from
which the juxtamembrane domain affects basolateral
localization of ADAM10 in epithelial cells [28]. In neu-
rones, the SH3 binding domains direct ADAM10 via
binding to synapse-associated protein-97 to the post-
synaptic membrane [29].
In 1997, the gene locus for ADAM10 was matched
to chromosome 15 in humans (15q21.3-q23) and chro-
mosome 9 in mice [30,31]. Subsequently, it took
8 years to achieve further gene structure analysis and
potential identification of transcription factor binding
sites [32]. We now know that the human, mouse and
rat genes, which comprise  160 kb, include a highly
homologous sequence within the first 500 bp upstream
of either translation initiation site. Deletion analysis
defined nucleotides )508 to )300 bp as the human
core promoter. This promoter was also identified as a
TATA-less promoter with functional binding sites for
Sp1, USF and retinoic acid receptors [32,33]. The func-
tional promoter of  2 kb displayed activity in various
human cell lines, such as HEK293, HepG2 or
SH-SY5Y, which reflects the ubiquitous basal expres-
sion of the endogenous ADAM10.

Single nucleotide polymorphism analyses of the pro-
moter region of 104 AD patients versus control
patients (n = 84) did not lead to significant statistical
differences [32]. In addition, an independent recent
study, genotyping 27 single nucleotide polymorphisms
covering the entire gene for ADAM10 in a larger
cohort of patients (n = 438 AD; n = 290 control),
revealed no single-marker or haplotypic association
with the disease [34]. This indicated that the gene for
ADAM10 probably does not constitute a major risk
with regard to AD. Nevertheless, a very recent study
of 1439 DNAs from 436 multiplex AD families yielded
significant evidence for an association of AD with the
metalloproteinase with respect to two mutations:
Q170H and R181G [35]. Both mutations are located
close to the cysteine switch within the prodomain and
the proprotein convertase recognition site (Fig. 1),
which explains their strong impact on enzyme func-
tionality: Chinese hamster ovary cells stably overex-
pressing mutated ADAM10 showed strongly
attenuated a-secretase activity [35]. Although both
mutations are rare (segregation in seven AD families
out of 1004) and are only partially penetrant, these
results give support to the hypothesis that the human
gene for ADAM10 plays a role in the aetiology of
AD.
ADAM10 and tumour necrosis factor a
(TACE): the ill-matched couple
Three members of the ADAM family have been shown
to act as a-secretase [8,36,37]: ADAM9, ADAM10 and

ADAM17 (TACE). Overexpression of ADAM9 has
been reported to increase the basal and protein kinase
C (PKC) dependent APPs-a release [36], although the
purified enzyme failed to cleave a synthetic peptide at
the major a-secretase cleavage-site [17]. Additionally,
mice lacking ADAM9 revealed no differences in the
production of the a-secretase cleavage product of APP
[38]. The impact of ADAM9 promoter polymorphism
on sporadic AD, which has been described recently
[39], might therefore rely on a more indirect mecha-
nism: ADAM9 has been shown to proteolytically pro-
cess ADAM10 [40–42]. By contrast to ADAM9,
ADAM10 was found to have constitutive and regu-
lated a-secretase activity as well as many other proper-
ties expected for the a-secretase [8,10]. Moreover,
in situ hybridization analysis in human cortical neuro-
nes provided evidence for the coexpression of APP
with ADAM10, suggesting that this proteinase is most
likely the physiologically relevant a-secretase [4].
Finally, experiments performed with ADAM17
(TACE)-deficient cells indicated a participation of
TACE in the regulated, PKC-stimulated [37,43] and
the constitutive a-secretase pathway [44,45]. To our
knowledge, there are no published reports about
TACE acting as an in vivo APP-sheddase in transgenic
mice, although TACE-positive neurones are found to
colocalize with amyloid plaques in AD brains support-
ing its role as an a-secretase [46].
On the basis of these results, it can be concluded
that ADAM10 and TACE are the major sheddases

that balance the b-site amyloid precursor protein cleav-
ing enzyme-driven generation of Ab peptides. This is
consistent with the close structural relationship of both
metalloproteinases: although TACE of human origin
has  30% amino acid identity relative to bovine
ADAM10, it only shows  15% identity with
ADAM9 [47]. Additionally, only those two ADAMs
lack the RX(6)DLPEFa(9)b(1) integrin binding motif,
which is contained in the other members of the pro-
teinase family [48]. Nevertheless, there are significant
differences between ADAM10 and TACE that
K. Endres and F. Fahrenholz Upregulation of ADAM10
FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS 1587
probably allow a specific modulation of one of them
for therapeutic approaches. TACE not only differs in
the consensus sequence of its disintegrin domain from
ADAM10 or by including a Crambin-like domain [47],
but also in its regulation. Several studies have
described the treatment of cellular cultures with a dis-
tinct outcome for either TACE or ADAM10 activity:
for example, incubation with phorbol 12-myristate 13-
acetate increased the turnover of TACE in Jurkat cells
[49] and diminished the amount of mature TACE in
HEK293 as well as in SH-SY5Y cells [45]. Interest-
ingly, these cell lines did not show altered amounts of
ADAM10, suggesting a significant difference in the cel-
lular stability of the mature enzyme forms after treat-
ment with 4b-phorbol 12-myristate 13-acetate [45]. In
addition, ADAM10 and TACE vary in their reaction
to cellular differentiation by retinoic acid [50,51] and

active site determinants of substrate recognition [52].
ADAM10: not particular about its
substrates?
For the enzyme ADAM10, more than 40 substrates
have been identified that belong to three different classes
of membrane bound proteins [53]. Most of them are
type I transmembrane proteins such as APP [8], APP-
like protein 2 [50] or the receptor for glycosylation end
products [54,55]. Type II transmembrane proteins such
as the apoptosis-inducing Fas ligand [56,57] or Bri2 [58]
have also been reported to be shed by ADAM10. Addi-
tionally, at least three glycosylphosphatidylinositol
(GPI)-anchored proteins are candidate substrates for
ADAM10: the metastasis-associated protein C4.4A was
characterized by a proteome technique as a substrate of
ADAM10 [59]. Furthermore, the GPI-anchored neuro-
nal guidance molecule ephrin A5 is cleaved by
ADAM10 upon binding to its receptor EphA3, leading
to termination of the receptor–ligand interaction [60].
Third, from cell culture experiments, the prion protein
PrP
c
was suggested to be processed by ADAM10 [40,61]
and the abundance of the PrP cleavage product C1 was
associated with mature ADAM10 within a small set of
human cerebral cortex samples [62]. However, in vivo
overexpression of ADAM10 in mice reduced all cellular
prion protein species instead of generating enhanced
amounts of cleavage products [63].
The substrates of ADAM10 show a

wide range of cellular function
ADAM10 cleaves proteins that affect cell migration
(N-cadherin [64]; transmembrane chemokines [65]) and
cell proliferation (CXCL16 [66–68]). It also sheds pro-
teins with functions in either the immune system (low
affinity immunoglobulin E receptor [69,70]; vascular
endothelial cadherin [71]) or in cell signalling (Delta
[72]; Notch [73]). Most effects, provoked by ADAM10
shedding activity, have been associated with the huge
N-terminal ectodomains of the substrates of ADAM10
that are released into the intercellular fluid upon cleav-
age. However, some effects have clearly been matched
to the intracellular domains of the substrates: ectodo-
main shedding by ADAM10 is followed by regulated
intramembrane proteolysis. After cleavage of the
Notch receptor by ADAM10, c-secretase releases a
small intracellular part of Notch, which then translo-
cates to the nucleus and acts as a transcription factor
[74–76]. With regard to Bri2, the ADAM10-derived
cleavage is followed by signal peptide peptidase-like
protease activity, also resulting in the release of a small
Bri2 fragment into the cell body [58].
In summary, ADAM10 has a repertoire of different
protein substrates hampering the development of ther-
apeutic strategies that target specifically APP by
ADAM10. However, not all substrates described as
being cleaved in the in vitro system have been con-
firmed in vivo. Mutagenesis experiments have depicted
at least three residues in the S1¢ pocket of ADAM10
that strongly influence substrate specificity and also

limit the number of substrates [52]. Additional interac-
tions of ADAM10 noncatalytical domains with the
substrate or with adaptor molecules, as previously
described for the recognition of ephrins [60], also
appear to be important for targeting ADAM10 to a
distinct substrate in the physiological context.
Regulators of ADAM10 expression and
catalytical activity
Because of the above-mentioned involvement of
ADAM10 in a wide range of cellular functions, it is
obvious to consider its therapeutic potential in various
diseases such as cancer or AD. ADAM10 has been
shown to cleave tumour-associated substrates such as
MICA [77] or C4.4A [59] and to be linked to progres-
sion of certain cancer types such as prostate or breast
cancer [78–80]. Furthermore, it plays a role in metasta-
sis of human colon cancer cells [81]. Therefore, the
inhibition of ADAM10 might be helpful in cancer
treatment in certain contexts [82]. By contrast,
ADAM10 overexpression or activation in the brain
might be beneficial for the treatment of neurodegenera-
tive diseases, in particular AD: this progressive disor-
der of the brain goes ahead with the loss of synaptic
junctions and neuronal cells. For ADAM10 overex-
pressing mice, it has been demonstrated that cortical
Upregulation of ADAM10 K. Endres and F. Fahrenholz
1588 FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS
synaptogenesis is enhanced [11], long-term potentiation
deficiency in AD model mice is rescued [10] and learn-
ing, as well as memory, is positively influenced by

ADAM10 [83]. Studies with the dominant negative
form of ADAM10 in a mouse model of AD revealed
that the enzymatic activity of ADAM10 is required to
counteract cognitive deficits [12]. In addition, axonal
guidance is conveyed by the metalloproteinase, as has
been shown for retinal and peripheral axons [84,85],
and ADAM10 regulates axon withdrawal by ephrin
cleavage [60,86].
It remains a matter of controversy as to whether
there is a substantial decline of neuronal ADAM10 in
ageing or in the pathological context: healthy, ageing
human fibroblasts did not reveal lowered amounts of
ADAM10 during senescence [87], although its specific
cleavage product APPs-a was decreased. Another
study demonstrated ADAM10 mRNA to be upregulat-
ed in cases of presenile dementia but to be downregu-
lated in the brain of AD patients [4]. A decrease for
ADAM10 and APPs-a was confirmed in human plate-
lets [88,89] as well as for APPs-a in the cerebrospinal
fluid of AD patients. Additionally, a recent study
revealed that colocalization of ADAM10 and one of
its potential regulators (i.e. nardilysin) is reduced in
AD compared to healthy aged brains [90].
With regard to these reports and to studies with
ADAM10 overexpression in a mouse AD model [10],
in principal, the enhancement of ADAM10 activity
and ⁄ or amount in the patient’s brain appears to be
valuable. How can this be achieved? Different
approaches appear to be promising, such as interfering
with the transcription ⁄ translation of ADAM10 or reg-

ulating its enzymatic capacity by influencing the mem-
brane physiology or via protein interactions (Fig. 2).
A first point of intervention within the biosynthetic
pathway of ADAM10 is provided by directly interfer-
ing with the expression of the gene for ADAM10: the
promoter region of the gene for ADAM10 has been
characterized in detail [32] and in silico analyses have
provided a multitude of transcription factor binding
sites. One of the putative binding sites for retinoic acid
receptors located at )302 and )203 bp has been dem-
onstrated to be functional by electrophoretic mobility
shift assay, promoter assays and APPs-a secretion in
human neuronal cells [32,50]. In addition, acitretin,
which is an accredited synthetic retinoid drug, lowered
Ab peptide generation in AD model mice and
enhanced APPs-a secretion [33]. Acitretin, which is
already used in the long-term treatment of patients suf-
fering from skin diseases withdraws all-trans retinoic
acid from its cellular retinoic acid binding protein and
makes it available for activating the corresponding
nuclear receptors. In the case of ADAM10 regulation,
cell culture studies with a variety of ligands for nuclear
receptors narrowed the receptors involved down
to a nonpermissive retinoic acid receptor–retinoid
X receptor heterodimer [33].
Another approach is offered by targeting the nascent
ADAM10 molecules during maturation within the cell.
Enhancement of the expression of a proprotein conver-
tase such as furin will increase ADAM10 maturation
Fig. 2. ADAM10 bears several points of vantage for its regulation.

For regulating the amount or catalytic activity of ADAM10, different
approaches such as interfering with membrane composition or pro-
teolytical processing of the proteinase itself are conceivable. In
addition, protein interaction partners such as TIMPs, tetraspanins
or reversion-inducing cysteine-rich protein with Kazal motifs (RECK)
modify the enzymatic property of ADAM10. GPCR-mediated cellular
signalling has been described for PACAP binding to PAC1 and tran-
scription factor based induction of gene expression (e.g. via retinoid
acid receptors) also contributes to ADAM10 activity within the cell.
Electrophoretic mobility shift assay experiments and application of
nuclear receptor ligands to the human neuroblastoma cell line
SH-SY5Y have identified important functional binding sites for non-
permissive retinoic acid receptor–retinoid X receptor heterodimers
at posititons )302 and ⁄ or )203 bp [32,33]. These can be directly
stimulated by addition of all-trans retinoic acid (atRA) or indirectly
by acitretin, liberating all-trans retinoic acid from cellular retinoic
acid binding protein. Pathways or molecules positively influencing
ADAM10 activity are indicated by a ‘+’ symbol, those with an
inhibitory effect by a ‘)’ symbol and those with an unknown
outcome by a ‘?’ symbol.
K. Endres and F. Fahrenholz Upregulation of ADAM10
FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS 1589
and a-secretase activity [21]. A further cleavage of
ADAM10 has been described in close proximity to
and within its transmembrane domain [40–42]. This is
a result of metalloproteinases ADAM9 and 15 acting
on ADAM10 to release a soluble sADAM10 from the
cell surface. sADAM10 was incapable of shedding cell-
associated amyloid precursor protein [42], whereas it
cleaved a synthetic peptide substrate [41,42] and

endogenous prion protein in cell culture experiments
[40]. Because it is still unclear whether soluble
ADAM10 and the transmembrane variant cleave the
same substrates and whether they have the same cata-
lytic properties in vivo, this type of regulation has to
be elucidated further. Acetylcholine esterase inhibitors,
which are already used in the symptomatic treatment
of AD, enhance the transport of ADAM10 to the cell
surface and the non-amylodogenic cleavage of APP
[91,92].
ADAM10 has also been shown to be regulated by
the lipid composition of the plasma membrane. While
cholesterol depletion enhanced its activity [93,94],
targeting ADAM10 via an artificial GPI-anchor to
cholesterol-rich domains inhibited its enzymatic func-
tion [95]. In human cells, the amount and activity of
ADAM10 was enhanced by statin application [93].
However, the outcomes of clinical trials with the chol-
esterol lowering statins are not unambiguous: several
studies have reported a protective effect of statins
against AD [96,97], although this could not be con-
firmed in others [98,99]. Nevertheless, in the prospec-
tive, population-based Rotterdam study comprising
 7000 participants, the use of statins was associated
with a lower risk of AD [100], preserving the hope of
a therapeutic value for statins in AD therapy. Further
evidence for lipids acting as modulators of a-secretase
activity is provided by a study demonstrating that type
III secretory phospholipase A and arachidonic acid
increased APPs-a production most likely by enhancing

substrate availability at the cell surface [101].
Another approach to activate ADAM10 could rest on
noncovalent protein interaction partners of ADAM10.
The tissue inhibitors of metalloproteinases 1 and 3 have
been shown to inhibit ADAM10 in vitro [102] and
the reversion-inducing cysteine-rich protein with Kazal
motifs also comprises a physiological ADAM10 inhibi-
tor [103]. By contrast, for the N-arginine dibasic
convertase (nardilysin), an activating property for
ADAM10-mediated APP a-secretase cleavage and
tumour necrosis factor a cleavage has been reported
[104,105]. The same holds true for the tetraspanins:
tetraspanin 12 increases maturation and activity of
ADAM10 [22] and ADAM10 has been suggested as a
component of the ‘tetraspanin web’ [106], which also
scaffolds heterotrimeric G protein-coupled receptors
(GPCRs) [107]. For the development of drugs interact-
ing with those proteins and thereby modulating
ADAM10 activity, further studies are necessary.
An appropriate strategy for targeting ADAM10 is
presented by directly stimulating the ADAM10 activity
by ligands of GPCRs. For example, the GPCR ligands
LPA and bombesin induced ADAM10-driven epider-
mal growth factor receptor transactivation [108] and
shedding of the thyrotropin receptor by ADAM10 was
mediated by its ligand thyrotropin [109]. At least in
cell culture, the a-secretase cleavage of APP is induc-
ible by the neuropeptide pituitary adenylate cyclase-
activating peptide (PACAP), which involves signalling
via mitogen-activated protein kinase and phosphatidyl-

inositol 3-kinase [110]. These results are of special
interest because the neuropeptide PACAP offers the
opportunity of locally activating the PAC1 receptor
and a-secretase in the brain. This also holds true for
the serotonin 5-hydroxytryptamine (5-HT4) receptor,
which increases memory and learning: the 5-HT4(e)
receptor isoform induced a-secretase activity by the
cAMP-regulated guanine exchange factor Epac and
the small GTPase Rac [111,112]. This recently led to
synthesis and evaluation of novel 5-HT4-agonists; two
of them increased APPs-a production in the cortex
and hippocampus of mice and exhibited neuroprotec-
tive properties [113].
Therefore, GPCR ligands offer an interesting oppor-
tunity in regulating ADAM10, even if the signalling
pathways have not yet been elucidated in every detail.
Another signalling pathway regulating ADAM10 activ-
ity is connected with the PKC: in various in vitro stud-
ies, it has been demonstrated that PKC or certain
isoforms of PKC stimulate the a-secretase [114–116]
(for the role of PKC in AD, see [117]) and this has
been confirmed in AD model mice (e.g. bryostatin 1)
[118].
ADAM10 as target for AD therapy:
lessons learned from transgenic mice
In summary, several independent strategies for enhanc-
ing the amount or the catalytic activity of ADAM10
have been performed or are conceivable. The crucial
question remaining is whether there are side effects
connected with enhanced ADAM10 activity in the

brain or in peripheral structures. ADAM10 mono-
transgenic mice with a permanent neuronal overexpres-
sion of ADAM10 to various extent were inconspicuous
in morphology, breeding and in daily handling [10].
This indicates that, by overexpression of ADAM10 in
the brain, the homeostasis of the entire organism is
Upregulation of ADAM10 K. Endres and F. Fahrenholz
1590 FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS
not grossly affected. A more detailed behavioural
examination showed that ADAM10 moderately over-
expressing mice performed similar to controls with
respect to basal activity, exploration and anxiety. In
the Morris water maze hidden platform task, however,
ADAM10 mono-transgenic mice showed thigmotaxis
with floating behaviour, indicating differences in moti-
vation [83]. Therefore, with respect to learning and
memory, mono-transgenic ADAM10 mice displayed
no specific phenotype. By contrast, overexpression of
ADAM10 in an AD mouse model with mutated
human APP created bi-transgenic mice with a clear
improvement of memory and alleviation of learning
deficits [10].
A recent microarray study [119] revealed that there
was only a moderate alteration of gene expression in
moderately ADAM10 overexpressing adult mice.
Genes coding for pro-inflammatory or pro-apoptotic
proteins were not over-represented among differentially
regulated genes and, indeed, a decrease of inflamma-
tion markers was observed. ADAM10 participates also
in the activation of Notch1 signalling by cleaving the

extracellular portion of this receptor upon ligand bind-
ing. Young ADAM10 transgenic mice at postnatal day
15 showed a 40% induction of expression of the gene
for Hes5, whereas a 50% reduction in mice overex-
pressing the dominant negative variant of the enzyme
was reported [119]. Nevertheless, in adult mice, no
significant effects with respect to the amount of
Notch1 target gene Hes5 mRNA were obtained, sug-
gesting an attenuation of the signalling cascade during
ageing. Because ADAM10-based AD therapy will take
place in elderly people, interference with this important
developmental signalling pathway does not appear to
hamper such an approach.
Regarding prion diseases, upregulation of ADAM10
might also be beneficial: the reduction of all species of
the prion protein in ADAM10 overexpressing mice
was accompanied by a prolonged survival time of the
mice after Scrapie infection [63]. In addition, Akt
phosphorylation as a marker for survival signals in
neuronal cells [120] was not affected in ADAM10
moderately overexpressing mice [121]. Furthermore,
the thickness of the myelin sheath was not altered by
ADAM10 overexpression, demonstrating that neuregu-
lin-1 acting as a modulator of this developmental event
is not a substrate of ADAM10 [121]. In mice charac-
terized by high levels of overexpressed ADAM10, how-
ever, phosphorylation of Akt was reduced to  50%
compared to wild-type mice and tomacula-like struc-
tures (i.e. local myelin thickenings) were observed
[121]. In addition, mice with high ADAM10 overex-

pression showed more seizures and stronger neuronal
damage and inflammation than wild-type mice upon
kainate treatment [122]. By contrast, in the presence of
its substrate APP in doses exceeding the endogenous
level, ADAM10 revealed a protective effect [122].
If we consider all of the results obtained concerning
increased ADAM10 activity in vivo, it can be con-
cluded that this approach might be a valuable alterna-
tive to other strategies, such as the inhibition of b-or
c-secretase or immunization, for the treatment of AD.
However, a-secretase activation must be moderate and
closely monitored.
Acknowledgements
The authors’ own work is supported by the Federal
Ministry of Education and Research (BMBF) in the
Framework of the National Genome Research Network
(NGFN), Fo
¨
rderkennzeichen FKZ01GS08130.
References
1 Chantry A, Gregson NA & Glynn P (1989) A novel
metalloproteinase associated with brain myelin
membranes. Isolation and characterization. J Biol
Chem 264, 21603–21607.
2 Chantry A, Gregson N & Glynn P (1992) Degradation
of myelin basic protein by a membrane-associated
metalloprotease: neural distribution of the enzyme.
Neurochem Res 17, 861–867.
3 Howard L, Lu X, Mitchell S, Griffiths S & Glynn P
(1996) Molecular cloning of MADM: a catalytically

active mammalian disintegrin-metalloprotease
expressed in various cell types. Biochem J 317, 45–50.
4 Marcinkiewicz M & Seidah NG (2000) Coordinated
expression of beta-amyloid precursor protein and the
putative beta-secretase BACE and alpha-secretase
ADAM10 in mouse and human brain. J Neurochem
75, 2133–2143.
5 Karkkainen I, Rybnikova E, Pelto-Huikko M &
Huovila AP (2000) Metalloprotease-disintegrin
(ADAM) genes are widely and differentially expressed
in the adult CNS. Mol Cell Neurosci 15, 547–560.
6 Dallas DJ, Genever PG, Patton AJ, Millichip MI,
McKie N & Skerry TM (1999) Localization of
ADAM10 and Notch receptors in bone. Bone 25 , 9–15.
7 McKie N, Edwards T, Dallas DJ, Houghton A, Stringer
B, Graham R, Russell G & Croucher PI (1997) Expres-
sion of members of a novel membrane linked metallo-
proteinase family (ADAM) in human articular
chondrocytes. Biochem Biophys Res Commun 230,
335–339.
8 Lammich S, Kojro E, Postina R, Gilbert S, Pfeiffer R,
Jasionowski M, Haass C & Fahrenholz F (1999)
Constitutive and regulated alpha-secretase cleavage of
K. Endres and F. Fahrenholz Upregulation of ADAM10
FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS 1591
Alzheimer’s amyloid precursor protein by a disintegrin
metalloprotease. Proc Natl Acad Sci U S A 96, 3922–
3927.
9 Roberts SB, Ripellino JA, Ingalls KM, Robakis NK &
Felsenstein KM (1994) Non-amyloidogenic cleavage of

the beta-amyloid precursor protein by an integral
membrane metalloendopeptidase. J Biol Chem 269,
3111–3116.
10 Postina R, Schroeder A, Dewachter I, Bohl J, Schmitt
U, Kojro E, Prinzen C, Endres K, Hiemke C, Blessing
M et al. (2004) A disintegrin-metalloproteinase pre-
vents amyloid plaque formation and hippocampal
defects in an Alzheimer disease mouse model. J Clin
Invest 113, 1456–1464.
11 Bell KF, Zheng L, Fahrenholz F & Cuello AC (2008)
ADAM-10 over-expression increases cortical synapto-
genesis. Neurobiol Aging 29, 554–565.
12 Schroeder A, Fahrenholz F & Schmitt U (2009) Effect of
a dominant-negative form of ADAM10 in a mouse
model of Alzheimer’s disease. J Alzheimers Dis 16, 309–
314.
13 Hooper NM & Turner AJ (2002) The search for alpha-
secretase and its potential as a therapeutic approach to
Alzheimer s disease. Curr Med Chem 9, 1107–1119.
14 Fahrenholz F & Postina R (2006) Alpha-secretase acti-
vation – an approach to Alzheimer’s disease therapy.
Neurodegener Dis 3, 255–261.
15 Fahrenholz F (2007) Alpha-secretase as a therapeutic
target. Curr. Alzheimer Res. 4, 412–417.
16 Anders A, Gilbert S, Garten W, Postina R &
Fahrenholz F (2001) Regulation of the alpha-secretase
ADAM10 by its prodomain and proprotein converta-
ses. FASEB J 15, 1837–1839.
17 Roghani M, Becherer JD, Moss ML, Atherton RE,
Erdjument-Bromage H, Arribas J, Blackburn RK,

Weskamp G, Tempst P & Blobel CP (1999) Metallo-
protease-disintegrin MDC9: intracellular maturation
and catalytic activity. J Biol Chem 274, 3531–3540.
18 Lum L, Reid MS & Blobel CP (1998) Intracellular
maturation of the mouse metalloprotease disintegrin
MDC15. J Biol Chem 273, 26236–26247.
19 Loechel F, Gilpin BJ, Engvall E, Albrechtsen R & Wewer
UM (1998) Human ADAM 12 (meltrin alpha) is an
active metalloprotease. J Biol Chem 273, 16993–16997.
20 Moss ML, Bomar M, Liu Q, Sage H, Dempsey P,
Lenhart PM, Gillispie PA, Stoeck A, Wildeboer D,
Bartsch JW et al. (2007) The ADAM10 prodomain is a
specific inhibitor of ADAM10 proteolytic activity and
inhibits cellular shedding events. J Biol Chem 282,
35712–35721.
21 Hwang EM, Kim SK, Sohn JH, Lee JY, Kim Y, Kim
YS & Mook-Jung I (2006) Furin is an endogenous
regulator of alpha-secretase associated APP processing.
Biochem Biophys Res Commun 349, 654–659.
22 Xu D, Sharma C & Hemler ME (2009) Tetraspanin12
regulates ADAM10-dependent cleavage of amyloid
precursor protein. FASEB J 23, 3674–3681.
23 Fahrenholz F, Gilbert S, Kojro E, Lammich S &
Postina R (2000) Alpha-secretase activity of the disin-
tegrin metalloprotease ADAM 10. Influences of
domain structure. Ann N Y Acad Sci 920, 215–222.
24 Escrevente C, Morais VA, Keller S, Soares CM,
Altevogt P & Costa J (2008) Functional role of N-gly-
cosylation from ADAM10 in processing, localization
and activity of the enzyme. Biochim Biophys Acta

1780, 905–913.
25 Horiuchi K, Le GS, Schulte M, Yamaguchi T, Reiss
K, Murphy G, Toyama Y, Hartmann D, Saftig P &
Blobel CP (2007) Substrate selectivity of epidermal
growth factor-receptor ligand sheddases and their regu-
lation by phorbol esters and calcium influx. Mol Biol
Cell 18, 176–188.
26 Nagano O, Murakami D, Hartmann D, De SB, Saftig
P, Iwatsubo T, Nakajima M, Shinohara M & Saya H
(2004) Cell-matrix interaction via CD44 is indepen-
dently regulated by different metalloproteinases acti-
vated in response to extracellular Ca(2+) influx and
PKC activation. J Cell Biol 165 , 893–902.
27 Sanderson MP, Erickson SN, Gough PJ, Garton KJ,
Wille PT, Raines EW, Dunbar AJ & Dempsey PJ
(2005) ADAM10 mediates ectodomain shedding of the
betacellulin precursor activated by p-aminophenylmer-
curic acetate and extracellular calcium influx. J Biol
Chem 280, 1826–1837.
28 Wild-Bode C, Fellerer K, Kugler J, Haass C & Capell
A (2006) A basolateral sorting signal directs ADAM10
to adherens junctions and is required for its function
in cell migration. J Biol Chem 281, 23824–23829.
29 Marcello E, Gardoni F, Mauceri D, Romorini S,
Jeromin A, Epis R, Borroni B, Cattabeni F, Sala C,
Padovani A et al. (2007) Synapse-associated protein-97
mediates alpha-secretase ADAM10 trafficking and pro-
motes its activity. J Neurosci 27 , 1682–1691.
30 Yamazaki K, Mizui Y, Sagane K & Tanaka I (1997)
Assignment of a disintegrin and metalloproteinase

domain 10 (Adam10) gene to mouse chromosome 9.
Genomics 46, 528–529.
31 Yamazaki K, Mizui Y & Tanaka I (1997) Radiation
hybrid mapping of human ADAM10 gene to chromo-
some 15. Genomics 45, 457–459.
32 Prinzen C, Muller U, Endres K, Fahrenholz F & Posti-
na R (2005) Genomic structure and functional charac-
terization of the human ADAM10 promoter. FASEB J
19, 1522–1524.
33 Tippmann F, Hundt J, Schneider A, Endres K &
Fahrenholz F (2009) Up-regulation of the alpha-
secretase ADAM10 by retinoic acid receptors and
acitretin. FASEB J 23, 1643–1654.
Upregulation of ADAM10 K. Endres and F. Fahrenholz
1592 FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS
34 Laws SM, Eckart K, Friedrich P, Kurz A, Forstl H &
Riemenschneider M (2009) Lack of evidence to sup-
port the association of polymorphisms within the
alpha- and beta-secretase genes (ADAM10 ⁄ BACE1)
with Alzheimer’s disease. Neurobiol Aging doi:10.1016/
j.neurobiolaging.2009.02.023.
35 Kim M, Suh J, Romano D, Truong MH, Mullin K,
Hooli B, Norton D, Tesco G, Elliott K, Wagner SL
et al. (2009) Potential late-onset Alzheimer’s disease-
associated mutations in the ADAM10 gene attenuate
{alpha}-secretase activity. Hum Mol Genet 18 , 3987–
3996.
36 Koike H, Tomioka S, Sorimachi H, Saido TC,
Maruyama K, Okuyama A, Fujisawa-Sehara A, Ohno
S, Suzuki K & Ishiura S (1999) Membrane-anchored

metalloprotease MDC9 has an alpha-secretase activity
responsible for processing the amyloid precursor pro-
tein. Biochem J 343, 371–375.
37 Buxbaum JD, Liu KN, Luo Y, Slack JL, Stocking
KL, Peschon JJ, Johnson RS, Castner BJ, Cerretti DP
& Black RA (1998) Evidence that tumor necrosis fac-
tor alpha converting enzyme is involved in regulated
alpha-secretase cleavage of the Alzheimer amyloid pro-
tein precursor. J Biol Chem 273, 27765–27767.
38 Weskamp G, Cai H, Brodie TA, Higashyama S,
Manova K, Ludwig T & Blobel CP (2002) Mice lack-
ing the metalloprotease-disintegrin MDC9 (ADAM9)
have no evident major abnormalities during develop-
ment or adult life. Mol Cell Biol 22 , 1537–1544.
39 Cong L & Jia J (2009) Promoter polymorphisms which
regulate ADAM9 transcription are protective against
sporadic Alzheimer’s disease. Neurobiol Aging
doi:10.1016/j.neurobiolaging.2009.01.001.
40 Cisse MA, Sunyach C, Lefranc-Jullien S, Postina R,
Vincent B & Checler F (2005) The disintegrin ADAM9
indirectly contributes to the physiological processing of
cellular prion by modulating ADAM10 activity. J Biol
Chem 280, 40624–40631.
41 Tousseyn T, Thathiah A, Jorissen E, Raemaekers T,
Konietzko U, Reiss K, Maes E, Snellinx A, Serneels L,
Nyabi O et al. (2009) ADAM10, the rate-limiting pro-
tease of regulated intramembrane proteolysis of Notch
and other proteins, is processed by ADAMS-9,
ADAMS-15, and the gamma-secretase. J Biol Chem
284, 11738–11747.

42 Parkin E & Harris B (2009) A disintegrin and metallo-
proteinase (ADAM)-mediated ectodomain shedding of
ADAM10. J Neurochem 108, 1464–1479.
43 Merlos-Suarez A, Fernandez-Larrea J, Reddy P, Base-
lga J & Arribas J (1998) Pro-tumor necrosis factor-
alpha processing activity is tightly controlled by a com-
ponent that does not affect notch processing. J Biol
Chem 273, 24955–24962.
44 Slack BE, Ma LK & Seah CC (2001) Constitutive
shedding of the amyloid precursor protein ectodomain
is up-regulated by tumour necrosis factor-alpha con-
verting enzyme. Biochem J 357, 787–794.
45 Endres K, Anders A, Kojro E, Gilbert S, Fahrenholz
F & Postina R (2003) Tumor necrosis factor-alpha
converting enzyme is processed by proprotein-conver-
tases to its mature form which is degraded upon
phorbol ester stimulation. Eur J Biochem 270, 2386–
2393.
46 Skovronsky DM, Fath S, Lee VM & Milla ME (2001)
Neuronal localization of the TNFalpha converting
enzyme (TACE) in brain tissue and its correlation to
amyloid plaques. J Neurobiol 49, 40–46.
47 Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack
JL, Wolfson MF, Castner BJ, Stocking KL, Reddy P,
Srinivasan S et al. (1997) A metalloproteinase disinte-
grin that releases tumour-necrosis factor-alpha from
cells. Nature 385, 729–733.
48 Eto K, Huet C, Tarui T, Kupriyanov S, Liu HZ,
Puzon-McLaughlin W, Zhang XP, Sheppard D, Engv-
all E & Takada Y (2002) Functional classification of

ADAMs based on a conserved motif for binding to
integrin alpha 9beta 1: implications for sperm-egg
binding and other cell interactions. J Biol Chem 277,
17804–17810.
49 Doedens JR & Black RA (2000) Stimulation-induced
down-regulation of tumor necrosis factor-alpha con-
verting enzyme. J Biol Chem 275, 14598–14607.
50 Endres K., Postina R, Schroeder A, Mueller U &
Fahrenholz F (2005) Shedding of the amyloid precur-
sor protein-like protein APLP2 by disintegrin-metallo-
proteinases. FEBS J 272, 5808–5820.
51 Holback S, Adlerz L, Gatsinzi T, Jacobsen KT &
Iverfeldt K (2008) PI3-K- and PKC-dependent up-reg-
ulation of APP processing enzymes by retinoic acid.
Biochem Biophys Res Commun 365, 298–303.
52 Caescu CI, Jeschke GR & Turk BE (2009) Active site
determinants of substrate recognition by the metallo-
proteinases TACE and ADAM10. Biochem J 424,
79–88.
53 Pruessmeyer J & Ludwig A (2009) The good, the bad
and the ugly substrates for ADAM10 and ADAM17 in
brain pathology, inflammation and cancer. Semin Cell
Dev Biol 20, 164–174.
54 Raucci A, Cugusi S, Antonelli A, Barabino SM,
Monti L, Bierhaus A, Reiss K, Saftig P & Bianchi ME
(2008) A soluble form of the receptor for advanced
glycation endproducts (RAGE) is produced by proteo-
lytic cleavage of the membrane-bound form by the shed-
dase a disintegrin and metalloprotease 10 (ADAM10).
FASEB J 22, 3716–3727.

55 Zhang L, Bukulin M, Kojro E, Roth A, Metz VV,
Fahrenholz F, Nawroth PP, Bierhaus A & Postina R
(2008) Receptor for advanced glycation end products is
subjected to protein ectodomain shedding by metallo-
proteinases. J Biol Chem 283, 35507–35516.
K. Endres and F. Fahrenholz Upregulation of ADAM10
FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS 1593
56 Kirkin V, Cahuzac N, Guardiola-Serrano F, Huault S,
Luckerath K, Friedmann E, Novac N, Wels WS,
Martoglio B, Hueber AO et al. (2007) The Fas ligand
intracellular domain is released by ADAM10 and
SPPL2a cleavage in T-cells. Cell Death Differ 14 ,
1678–1687.
57 Schulte M, Reiss K, Lettau M, Maretzky T, Ludwig
A, Hartmann D, De SB, Janssen O & Saftig P (2007)
ADAM10 regulates FasL cell surface expression and
modulates FasL-induced cytotoxicity and activation-
induced cell death. Cell Death Differ 14, 1040–1049.
58 Martin L, Fluhrer R, Reiss K, Kremmer E, Saftig P &
Haass C (2008) Regulated intramembrane proteolysis
of Bri2 (Itm2b) by ADAM10 and SPPL2a ⁄ SPPL2b. J
Biol Chem 283, 1644–1652.
59 Esselens CW, Malapeira J, Colome N, Moss M,
Canals F & Arribas J (2008) Metastasis-associated
C4.4A, a GPI-anchored protein cleaved by ADAM10
and ADAM17. Biol Chem 389, 1075–1084.
60 Janes PW, Saha N, Barton WA, Kolev MV, Wimmer-
Kleikamp SH, Nievergall E, Blobel CP, Himanen JP,
Lackmann M & Nikolov DB (2005) Adam meets Eph:
an ADAM substrate recognition module acts as a

molecular switch for ephrin cleavage in trans. Cell 123,
291–304.
61 Vincent B, Paitel E, Saftig P, Frobert Y, Hartmann D,
De SB, Grassi J, Lopez-Perez E & Checler F (2001)
The disintegrins ADAM10 and TACE contribute to
the constitutive and phorbol ester-regulated normal
cleavage of the cellular prion protein. J Biol Chem 276,
37743–37746.
62 Laffont-Proust I, Faucheux BA, Hassig R, Sazdovitch
V, Simon S, Grassi J, Hauw JJ, Moya KL & Haik S
(2005) The N-terminal cleavage of cellular prion pro-
tein in the human brain. FEBS Lett 579, 6333–6337.
63 Endres K, Mitteregger G, Kojro E, Kretzschmar H &
Fahrenholz F (2009) Influence of ADAM10 on prion
protein processing and scrapie infectiosity in vivo.
Neurobiol Dis 36, 233–241.
64 Kohutek ZA, diPierro CG, Redpath GT & Hussaini
IM (2009) ADAM-10-mediated N-cadherin cleavage
is protein kinase C-alpha dependent and
promotes glioblastoma cell migration. J Neurosci 29,
4605–4615.
65 Hundhausen C, Schulte A, Schulz B, Andrzejewski
MG, Schwarz N, von HP, Winter U, Paliga K, Reiss
K, Saftig P et al. (2007) Regulated shedding of trans-
membrane chemokines by the disintegrin and metallo-
proteinase 10 facilitates detachment of adherent
leukocytes. J Immunol 178, 8064–8072.
66 Gough PJ, Garton KJ, Wille PT, Rychlewski M,
Dempsey PJ & Raines EW (2004) A disintegrin and
metalloproteinase 10-mediated cleavage and shedding

regulates the cell surface expression of CXC chemokine
ligand 16. J Immunol 172, 3678–3685.
67 Schulte A, Schulz B, Andrzejewski MG, Hundhausen
C, Mletzko S, Achilles J, Reiss K, Paliga K, Weber C,
John SR et al. (2007) Sequential processing of the
transmembrane chemokines CX3CL1 and CXCL16 by
alpha- and gamma-secretases. Biochem Biophys Res
Commun 358, 233–240.
68 Schramme A, bdel-Bakky MS, Kampfer-Kolb N, Pfe-
ilschifter J & Gutwein P (2008) The role of CXCL16
and its processing metalloproteinases ADAM10 and
ADAM17 in the proliferation and migration of human
mesangial cells. Biochem Biophys Res Commun 370,
311–316.
69 Weskamp G, Ford JW, Sturgill J, Martin S, Docherty
AJ, Swendeman S, Broadway N, Hartmann D, Saftig
P, Umland S et al. (2006) ADAM10 is a principal
‘sheddase’ of the low-affinity immunoglobulin E recep-
tor CD23. Nat Immunol 7, 1293–1298.
70 Lemieux GA, Blumenkron F, Yeung N, Zhou P, Wil-
liams J, Grammer AC, Petrovich R, Lipsky PE, Moss
ML & Werb Z (2007) The low affinity IgE receptor
(CD23) is cleaved by the metalloproteinase ADAM10.
J Biol Chem 282, 14836–14844.
71 Schulz B, Pruessmeyer J, Maretzky T, Ludwig A,
Blobel CP, Saftig P & Reiss K (2008) ADAM10 regu-
lates endothelial permeability and T-Cell transmigra-
tion by proteolysis of vascular endothelial cadherin.
Circ Res 102
, 1192–1201.

72 Dyczynska E, Sun D, Yi H, Sehara-Fujisawa A, Blobel
CP & Zolkiewska A (2007) Proteolytic processing of
delta-like 1 by ADAM proteases. J Biol Chem 282,
436–444.
73 Tian L, Wu X, Chi C, Han M, Xu T & Zhuang Y
(2008) ADAM10 is essential for proteolytic activation
of Notch during thymocyte development. Int Immunol
20, 1181–1187.
74 Schroeter EH, Kisslinger JA & Kopan R (1998)
Notch-1 signalling requires ligand-induced proteolytic
release of intracellular domain. Nature 393, 382–386.
75 De SB, Annaert W, Cupers P, Saftig P, Craessaerts K,
Mumm JS, Schroeter EH, Schrijvers V, Wolfe MS,
Ray WJ et al. (1999) A presenilin-1-dependent gamma-
secretase-like protease mediates release of Notch intra-
cellular domain. Nature 398, 518–522.
76 Cagavi BE & Weinmaster G (2009) Selective use of
ADAM10 and ADAM17 in activation of Notch1
signaling. Mol Cell Biol 29, 5679–5695.
77 Waldhauer I, Goehlsdorf D, Gieseke F, Weinschenk T,
Wittenbrink M, Ludwig A, Stevanovic S, Rammensee
HG & Steinle A (2008) Tumor-associated MICA is
shed by ADAM proteases. Cancer Res 68, 6368–6376.
78 McCulloch DR, Akl P, Samaratunga H, Herington
AC & Odorico DM (2004) Expression of the disinte-
grin metalloprotease, ADAM-10, in prostate cancer
and its regulation by dihydrotestosterone, insulin-like
growth factor I, and epidermal growth factor in the
Upregulation of ADAM10 K. Endres and F. Fahrenholz
1594 FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS

prostate cancer cell model LNCaP. Clin Cancer Res
10, 314–323.
79 Arima T, Enokida H, Kubo H, Kagara I, Matsuda R,
Toki K, Nishimura H, Chiyomaru T, Tatarano S, Ide-
sako T et al. (2007) Nuclear translocation of ADAM-
10 contributes to the pathogenesis and progression of
human prostate cancer. Cancer Sci 98, 1720–1726.
80 Lendeckel U, Kohl J, Arndt M, Carl-McGrath S,
Donat H & Rocken C (2005) Increased expression of
ADAM family members in human breast cancer and
breast cancer cell lines. J Cancer Res Clin Oncol 131,
41–48.
81 Gavert N, Sheffer M, Raveh S, Spaderna S, Shtutman
M, Brabletz T, Barany F, Paty P, Notterman D,
Domany E et al. (2007) Expression of L1-CAM and
ADAM10 in human colon cancer cells induces metas-
tasis. Cancer Res 67, 7703–7712.
82 Moss ML, Stoeck A, Yan W & Dempsey PJ (2008)
ADAM10 as a target for anti-cancer therapy. Curr
Pharm Biotechnol 9, 2–8.
83 Schmitt U, Hiemke C, Fahrenholz F & Schroeder A
(2006) Over-expression of two different forms of the
alpha-secretase ADAM10 affects learning and memory
in mice. Behav Brain Res 175, 278–284.
84 Chen YY, Hehr CL, tkinson-Leadbeater K, Hocking
JC & McFarlane S (2007) Targeting of retinal axons
requires the metalloproteinase ADAM10. J Neurosci
27, 8448–8456.
85 Jangouk P, Dehmel T, Meyer Zu HG, Ludwig A, Leh-
mann HC & Kieseier BC (2009) Involvement of

ADAM10 in axonal outgrowth and myelination of the
peripheral nerve. Glia 57, 1765–1774.
86 Hattori M, Osterfield M & Flanagan JG (2000)
Regulated cleavage of a contact-mediated axon repel-
lent. Science 289, 1360–1365.
87 Kern A, Roempp B, Prager K, Walter J & Behl C
(2006) Down-regulation of endogenous amyloid pre-
cursor protein processing due to cellular aging. J Biol
Chem 281, 2405–2413.
88 Colciaghi F, Borroni B, Pastorino L, Marcello E,
Zimmermann M, Cattabeni F, Padovani A & Di LM
(2002) [alpha]-Secretase ADAM10 as well as
[alpha]APPs is reduced in platelets and CSF of Alzhei-
mer disease patients. Mol Med 8, 67–74.
89 Colciaghi F, Marcello E, Borroni B, Zimmermann M,
Caltagirone C, Cattabeni F, Padovani A & Di LM
(2004) Platelet APP, ADAM 10 and BACE alterations
in the early stages of Alzheimer disease. Neurology 62,
498–501.
90 Bernstein HG, Stricker R, Lendeckel U, Bertram I,
Dobrowolny H, Steiner J, Bogerts B & Reiser G
(2009) Reduced neuronal co-localisation of nardilysin
and the putative alpha-secretases ADAM10 and
ADAM17 in Alzheimer’s disease and Down syndrome
brains. Age (Dordr) 31, 11–25.
91 Yogev-Falach M, Amit T, Bar-Am O, Weinstock M &
Youdim MB (2002) Involvement of MAP kinase in the
regulation of amyloid precursor protein processing by
novel cholinesterase inhibitors derived from rasagiline.
FASEB J 16, 1674–1676.

92 Zimmermann M, Gardoni F, Marcello E, Colciaghi F,
Borroni B, Padovani A, Cattabeni F & Di LM (2004)
Acetylcholinesterase inhibitors increase ADAM10
activity by promoting its trafficking in neuroblastoma
cell lines. J Neurochem 90, 1489–1499.
93 Kojro E, Gimpl G, Lammich S, Marz W & Fahren-
holz F (2001) Low cholesterol stimulates the nonamy-
loidogenic pathway by its effect on the alpha -secretase
ADAM 10. Proc Natl Acad Sci U S A 98, 5815–5820.
94 Matthews V, Schuster B, Schutze S, Bussmeyer I,
Ludwig A, Hundhausen C, Sadowski T, Saftig P,
Hartmann D, Kallen KJ et al. (2003) Cellular choles-
terol depletion triggers shedding of the human interleu-
kin-6 receptor by ADAM10 and ADAM17 (TACE). J
Biol Chem 278, 38829–38839.
95 Harris B, Pereira I & Parkin E (2009) Targeting
ADAM10 to lipid rafts in neuroblastoma SH-SY5Y
cells impairs amyloidogenic processing of the amyloid
precursor protein. Brain Res 1296, 203–215.
96 Wolozin B, Kellman W, Ruosseau P, Celesia GG &
Siegel G (2000) Decreased prevalence of Alzheimer
disease associated with 3-hydroxy-3-methyglutaryl coen-
zyme A reductase inhibitors. Arch Neurol 57, 1439–
1443.
97 Rockwood K, Kirkland S, Hogan DB, MacKnight C,
Merry H, Verreault R, Wolfson C & McDowell I
(2002) Use of lipid-lowering agents, indication bias,
and the risk of dementia in community-dwelling elderly
people. Arch Neurol 59, 223–227.
98 Rea TD, Breitner JC, Psaty BM, Fitzpatrick AL, Lo-

pez OL, Newman AB, Hazzard WR, Zandi PP, Burke
GL, Lyketsos CG et al. (2005) Statin use and the risk
of incident dementia: the Cardiovascular Health Study.
Arch Neurol 62, 1047–1051.
99 Zandi PP, Sparks DL, Khachaturian AS, Tschanz J,
Norton M, Steinberg M, Welsh-Bohmer KA & Breit-
ner JC (2005) Do statins reduce risk of incident demen-
tia and Alzheimer disease? The Cache County Study
Arch Gen Psychiatry 62, 217–224.
100 Haag MD, Hofman A, Koudstaal PJ, Stricker BH &
Breteler MM (2009) Statins are associated with a
reduced risk of Alzheimer disease regardless of lipophi-
licity. The Rotterdam Study. J Neurol Neurosurg
Psychiatry 80, 13–17.
101 Yang X, Sheng W, He Y, Cui J, Haidekker MA, Sun
GY & Lee JC (2009) Secretory phospholipase A2 type
III enhances {alpha}-secretase-dependent amyloid
precursor protein processing through alterations in
membrane fluidity. J Lipid Res doi: 10.1194/
jlr.M002287.
K. Endres and F. Fahrenholz Upregulation of ADAM10
FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS 1595
102 Amour A, Knight CG, Webster A, Slocombe PM,
Stephens PE, Knauper V, Docherty AJ & Murphy G
(2000) The in vitro activity of ADAM-10 is inhibited
by TIMP-1 and TIMP-3. FEBS Lett 473, 275–279.
103 Muraguchi T, Takegami Y, Ohtsuka T, Kitajima S,
Chandana EP, Omura A, Miki T, Takahashi R,
Matsumoto N, Ludwig A et al. (2007) RECK modulates
Notch signaling during cortical neurogenesis by regulat-

ing ADAM10 activity. Nat Neurosci 10, 838–845.
104 Hiraoka Y, Yoshida K, Ohno M, Matsuoka T, Kita T
& Nishi E (2008) Ectodomain shedding of TNF-alpha
is enhanced by nardilysin via activation of ADAM
proteases. Biochem Biophys Res Commun 370, 154–158.
105 Hiraoka Y, Ohno M, Yoshida K, Okawa K, Tomimot-
o H, Kita T & Nishi E (2007) Enhancement of alpha-
secretase cleavage of amyloid precursor protein by a
metalloendopeptidase nardilysin. J Neurochem 102,
1595–1605.
106 Arduise C, Abache T, Li L, Billard M, Chabanon A,
Ludwig A, Mauduit P, Boucheix C, Rubinstein E &
Le NF (2008) Tetraspanins regulate ADAM10-medi-
ated cleavage of TNF-alpha and epidermal growth fac-
tor. J Immunol 181, 7002–7013.
107 Little KD, Hemler ME & Stipp CS (2004) Dynamic
regulation of a GPCR-tetraspanin-G protein complex
on intact cells: central role of CD81 in facilitating
GPR56-Galpha q ⁄ 11 association. Mol Biol Cell 15,
2375–2387.
108 Yan Y, Shirakabe K & Werb Z (2002) The metallopro-
tease Kuzbanian (ADAM10) mediates the transactiva-
tion of EGF receptor by G protein-coupled receptors.
J Cell Biol 158, 221–226.
109 Kaczur V, Puskas LG, Nagy ZU, Miled N, Rebai A,
Juhasz F, Kupihar Z, Zvara A, Hackler L Jr & Farid
NR (2007) Cleavage of the human thyrotropin receptor
by ADAM10 is regulated by thyrotropin. J Mol
Recognit 20, 392–404.
110 Kojro E, Postina R, Buro C, Meiringer C, Gehrig-

Burger K & Fahrenholz F (2006) The neuropeptide
PACAP promotes the alpha-secretase pathway for
processing the Alzheimer amyloid precursor protein.
FASEB J 20, 512–514.
111 Robert S, Maillet M, Morel E, Launay JM, Fischmei-
ster R, Mercken L & Lezoualc’h F (2005) Regulation
of the amyloid precursor protein ectodomain shedding
by the 5-HT4 receptor and Epac. FEBS Lett 579,
1136–1142.
112 Robert SJ, Zugaza JL, Fischmeister R, Gardier AM &
Lezoualc’h F (2001) The human serotonin 5-HT4
receptor regulates secretion of non-amyloidogenic
precursor protein. J Biol Chem 276, 44881–44888.
113 Russo O, Cachard-Chastel M, Riviere C, Giner M,
Soulier JL, Berthouze M, Richard T, Monti JP, Sicsic
S, Lezoualc’h F et al. (2009) Design, synthesis, and
biological evaluation of new 5-HT4 receptor agonists:
application as amyloid cascade modulators and poten-
tial therapeutic utility in Alzheimer’s disease. J Med
Chem 52, 2214–2225.
114 Gillespie SL, Golde TE & Younkin SG (1992) Secre-
tory processing of the Alzheimer amyloid beta ⁄ A4 pro-
tein precursor is increased by protein phosphorylation.
Biochem Biophys Res Commun 187, 1285–1290.
115 Hung AY & Selkoe DJ (1994) Selective ectodomain
phosphorylation and regulated cleavage of beta-amy-
loid precursor protein. EMBO J 13, 534–542.
116 Skovronsky DM, Moore DB, Milla ME, Doms RW &
Lee VM (2000) Protein kinase C-dependent alpha-
secretase competes with beta-secretase for cleavage of

amyloid-beta precursor protein in the trans-golgi
network. J Biol Chem 275, 2568–2575.
117 Alkon DL, Sun MK & Nelson TJ (2007) PKC signal-
ing deficits: a mechanistic hypothesis for the origins of
Alzheimer’s disease. Trends Pharmacol Sci 28, 51–60.
118 Etcheberrigaray R, Tan M, Dewachter I, Kuiperi C,
Van dA I, Wera S, Qiao L, Bank B, Nelson TJ,
Kozikowski AP et al. (2004) Therapeutic effects of
PKC activators in Alzheimer’s disease transgenic mice.
Proc Natl Acad Sci U S A 101, 11141–11146.
119 Prinzen C, Trumbach D, Wurst W, Endres K, Postina
R & Fahrenholz F (2009) Differential gene expression
in ADAM10 and mutant ADAM10 transgenic mice.
BMC Genomics 10, 66.
120 Datta SR, Brunet A & Greenberg ME (1999) Cellular
survival: a play in three Akts. Genes Dev 13, 2905–
2927.
121 Freese C, Garratt AN, Fahrenholz F & Endres K
(2009) The effects of alpha-secretase ADAM10 on
the proteolysis of neuregulin-1. FEBS J 276, 1568–
1580.
122 Clement AB, Hanstein R, Schroder A, Nagel H,
Endres K, Fahrenholz F & Behl C (2008) Effects of
neuron-specific ADAM10 modulation in an in vivo
model of acute excitotoxic stress. Neuroscience 152,
459–468.
Upregulation of ADAM10 K. Endres and F. Fahrenholz
1596 FEBS Journal 277 (2010) 1585–1596 ª 2010 The Authors Journal compilation ª 2010 FEBS

×