You are on page 1of 14

Journal of Pathology

J Pathol 2012; 226: 158–171 INVITED REVIEW


Published online in Wiley Online Library
(wileyonlinelibrary.com) DOI: 10.1002/path.3027

Cell–cell connectivity: desmosomes and disease


Matthew A Brooke,# Daniela Nitoiu# and David P Kelsell*
Centre for Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, London, UK

*Correspondence to: David P Kelsell, Centre for Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry,
Queen Mary University of London, 4 Newark Street, London E1 2AT, UK. e-mail: d.p.kelsell@qmul.ac.uk
# Joint first authors.

Abstract
Cell–cell connectivity is an absolute requirement for the correct functioning of cells, tissues and entire
organisms. At the level of the individual cell, direct cell–cell adherence and communication is mediated by the
intercellular junction complexes: desmosomes, adherens, tight and gap junctions. A broad spectrum of inherited,
infectious and auto-immune diseases can affect the proper function of intercellular junctions and result in either
diseases affecting specific individual tissues or widespread syndromic conditions. A particularly diverse group of
diseases result from direct or indirect disruption of desmosomes—a consequence of their importance in tissue
integrity, their extensive distribution, complex structure, and the wide variety of functions their components
accomplish. As a consequence, disruption of desmosomal assembly, structure or integrity disrupts not only their
intercellular adhesive function but also their functions in cell communication and regulation, leading to such
diverse pathologies as cardiomyopathy, epidermal and mucosal blistering, palmoplantar keratoderma, woolly hair,
keratosis, epidermolysis bullosa, ectodermal dysplasia and alopecia. Here, as well as describing the importance of
the other intercellular junctions, we focus primarily on the desmosome, its structure and its role in disease. We will
examine the various pathologies that result from impairment of desmosome function and thereby demonstrate
the importance of desmosomes to tissues and to the organism as a whole.
Copyright  2011 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Keywords: desmosome; cadherins; desmoplakin; hyperadhesion; proteases; pemphigus; cystatin A; ADAM17; ARVC

Received 23 September 2011; Revised 3 October 2011; Accepted 3 October 2011

No conflicts of interest were declared.

Introduction wide distribution and key roles in maintaining tissue


integrity and in cellular signalling mean that a broad
Connections between individual cells are a hallmark variety of diseases can result from their dysfunction or
of, and an absolute requirement for, multicellular life. dysregulation.
Connectivity in terms of intercellular communication
can be accomplished between distantly separated cells Non-desmosomal intercellular junctions in disease
within an organism by various means, such as ner- Adherens junctions can be grouped with desmosomes
vous transmission, hormonal signalling in endocrine or as ‘anchoring’ or ‘adhering’ junctions, whose role pri-
paracrine fashions, or through specialized fluids that marily features organizing and tethering the cytoskele-
function specifically to link distant cells, such as the ton (condensed actin filaments in the case of adherens
blood or lymph. Meanwhile, direct cell–cell connec- junctions, intermediate filaments for desmosomes) to
tions are mediated by intercellular junction complexes; the plasma membrane, and maintaining close physi-
a diverse group of organelles—including desmosomes, cal association between cells [1]. Sharing a kind of
adherens, tight and gap junctions—which facilitate structural homology with desmosomes, adherens junc-
adherence and communication between individual cells tions are assembled from: classical cadherins (such
and maintain the integrity of larger tissues. The impor- as E-, N- and P-cadherin)—calcium-dependent trans-
tance of intercellular junctions, as with many other membrane proteins extending out from the cell sur-
features of the human body, is most amply demon- face, which bind cadherins on adjacent cells, originally
strated by their involvement in a wide-ranging vari- named for the tissue in which they are were thought
ety of diseases, of genetic, auto-immune, cancerous to be mainly expressed (eg E-cadherin in epithelium,
and infectious aetiology. This review will examine the N-cadherin in neuronal tissue) [2]; armadillo proteins
importance of cell–cell connectivity by discussing the (such as β-catenin and plakoglobin (PG)—the lat-
basis of diseases involving the intercellular junctions. ter also in desmosomes), which bind to intracellular
Particular attention will be paid to desmosomes, whose domains of the cadherins and have important roles in
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 159

signalling pathways and adherence [3]; and cytoskele- with deafness (reviewed by Scott and Kelsell [11]).
tal adaptor proteins (such as α-catenin), which were Deafness as a result of connexin mutations (particu-
thought to provide a link between the armadillo pro- larly affecting Cx26) accounts for a major proportion
teins and the actin cytoskeleton until it was shown that of genetic non-syndromic hearing loss in many ethnic
α-catenin in complex with a cadherin and β-catenin groups [14,15].
cannot bind F-actin [4]. Diseases that result from muta- The primary role of the tight junctions is thought
tions in adherens junction components reflect their to be in the maintenance of the epithelial barrier,
wide distribution in many tissues. For example, defi- where they function to regulate the permeability of the
ciencies of P-cadherin have been reported in the related paracellular pathway between epithelial cells; tightly
conditions hypotrichosis with juvenile macular dystro- sealing the barrier to the point of high resistance in
phy (HJMD) [5] and ectodermal dysplasia, ectrodactyly tissues such as the bladder, whilst providing a more
and macular dystrophy (EEM syndrome) [6]. Both fea- permeable barrier—to facilitate water and nutrient
ture early hair loss and progressive degeneration of the absorption—in the intestinal epithelium, for example
central retina, with the latter accompanied by hypodon- [16]. The best characterized of the proteins that make
tia and limb defects such as hand or foot malforma- up tight junctions are the claudins, a transmembrane
tion, the affected bodily sites correlating with areas of protein family of at least 24 members, expressed in
strong P-cadherin expression during development [7]. tissue-specific combinations [17]. Although almost no
As well as its function in adherens junctions, β-catenin information is known about how claudins oligomerize
is also a crucial component of the Wnt signalling path- into the higher-order structures seen in tight junctions,
way, which regulates development and homeostasis via or now they adhere across the intercellular space, their
gene expression, cell growth, survival and polarity [8]. first extracellular loop has been shown to define to
Wnt pathway signalling is regulated by phosphoryla- which ions (and non-ionic solutes) a tight junction is
tion and subsequent degradation of β-catenin under permeable—defining its ionic charge selectivity [18].
normal circumstances, meaning that somatic mutations Proteins associated with claudins at the tight junction
in β-catenin itself, or in those proteins involved in include the cytoplasmic ZO-1, -2 and -3 family and
its phosphorylation or degradation [including Dishev- other transmembrane proteins, such as occludin and
elled, Axin, Adenomatous Polyposis Coli (APC) and tricellulin. The tight junction cytoplasmic plaque and
glycogen synthase kinase-3β] can lead to constitutive non-claudin transmembrane proteins are reviewed by
β-catenin activation and thus cancer development [8]. Schneeberger and Lynch [19]. Mutations in a vari-
Mutations in these proteins have been observed in a ety of the genes encoding the claudins constitute the
very wide variety of tumour types and are reviewed majority of tight junction-associated disease. CLDN16
well by Polakis [9] and Xueling and Xin [10], among (claudin 16) loss-of-function mutations, for example,
others. cause hypomagnesaemia, hypercalciuria and nephro-
Gap junctions serve to permit intercellular commu- calcinosis, as a result of impaired Ca2+ and Mg2+
nication, a function vital for controlling homeostasis reabsorbtion via cation-selective tight junctions in the
in organisms and for responses to external stimuli, renal tubule [20] [21], whereas mutations in CLDN19
which they accomplish by allowing the transfer of ions (claudin 19) can cause a similar syndrome accompa-
(including Ca2+ and Mg2+ ) and small molecules of nied by visual impairment [22]. Mutations in the asso-
<1 kDa (such as cAMP, cGMP and ATP) between ciated proteins ZO-2 [23] and tricellulin [24] can lead
cells [11]. Gap junctions consist of plaques of many to familial hypercholanaemia and non-syndromic deaf-
small channels, each the product of two hexameric ness, respectively.
hemi-channels on closely apposed cell membranes.
Each hemi-channel is a homo- or heteromeric hex-
amer made up of connexins (Cx), a protein family of
Desmosome structure and component function
21 members in humans, whose members are named Desmosomes not only provide mechanical stability but
according to their molecular mass (eg Cx26, weighing also facilitate cell–cell communication through sig-
approximately 26 kDa) [12]. Connexins are differen- nal transmission. Desmosomal structures have been
tially expressed in the human body, with multiple types observed in various tissue types that experience
expressed in any single tissue type. The range of disor- mechanical stress, such as the intestinal mucosa, gall-
ders—including skin disease, non-syndromic deafness, bladder, uterus and oviduct, liver, pancreas, stomach,
neuropathy, and syndromic diseases (affecting multiple salivary and thyroid glands and the epithelial cells of
tissue types) [13]—associated with Cx mutations illus- the nephron, but are most abundant in the skin and
trates their wide distribution and physiological impor- myocardium [25–28]. A primary function of desmo-
tance. Connexins’ widespread role is further demon- somes is the anchoring of cytoskeletal keratin interme-
strated by the fact that different mutations in the same diate filaments in the epidermis, desmin intermediate
connexin gene can lead to completely different phe- filaments in the heart and vimentin intermediate fila-
notypes, with either dominant or recessive inheritance. ments in meningeal cells and the follicular dendritic
For example, mutations in GJB3 (the gene encoding cells of lymph nodes to the cell membrane [29].
Cx31) can result in non-syndromic deafness, erythro- The desmosome’s structure was first observed by
keratoderma variabilis alone, or peripheral neuropathy the Italian pathologist Bizzozero (1864); its structure
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
160 MA Brooke et al

Figure 1. Desmosome structure. (A) EM of a young desmosome, which lacks a dense midline, connecting two adjacent cells in a
keratinocyte monolayer. (B) Schematic representation of desmosomal structure. Desmogleins (DSGs) and desmocollins (DSCs) constitute
the extracellular link between cells, and are responsible for the appearance of the dense midline; they form homo- and heterodimers,
whose intracellular tails bind the Armadillo family proteins plakoglobin (PG) and the plakophilins (PKPs), which serve to link cadherins to
the plakin protein desmoplakin (DSP), which in turn tethers desmosomes to the intermediate filament network.

has since been analysed by techniques such as elec-


tron microscopy (EM) to reveal a complex structure
and organisation. The desmosome divides into three
parallel identifiable zones, arranged symmetrically on
the cytoplasmic faces of the plasma membranes of bor-
dering cells and separated by the extracellular domain,
which in mature desmosomes is bisected by a dense
midline [30]. Each desmosomal plaque consists of
a thick outer dense plaque and a translucent inner
dense plaque [31]. The five major desmosomal com-
ponents are the desmosomal cadherins, represented Figure 2. Desmosomal cadherin expression in the epidermis. This
figure illustrates the differential distribution and relative expression
by desmogleins (DSG1-4) and desmocollins (DSC1- levels of the desmosomal cadherins—the DSGs and DSCs—in the
3), the armadillo family members, plakoglobin (PG) epidermis. The location and depth of blistering observed in diseases
and the plakophilins (PKP1-3), and the plakin linker such as pemphigus vulgaris and foliaceus reflects this distribution.
protein desmoplakin (DSP), which anchors the interme-
diate keratin filaments. The structure of a desmosome different tissues are biochemically and functionally dis-
is illustrated in Figure 1. tinct. The precise role for the tissue-specific expres-
sion patterns of desmosomal cadherins is not fully
Desmosomal cadherins and Ca2+ -mediated understood, but manipulation of desmosomal cad-
hyperadhesion herin expression suggests that tight regulation of their
Desmogleins (DSGs) and desmocollins (DSCs) are expression pattern is critical to tissue homeostasis [37].
transmembrane components that bridge adjacent cells Within the epidermis, these genes are differentially
and are embedded in the cytoplasmic plaques, and expressed as keratinocytes undergo terminal differ-
have been identified to form the dense midline seen in entiation [38] [28]: DSG1 and DSC1 are strongly
mature desmosomes. They share 30% amino acid iden- expressed in the granular and spinous layers, their
tity between each other and with classical cadherins levels decreasing in the lower levels of the epider-
[32], with DSC genes being more closely related to mis [39–41]; DSG2 and DSC2 are expressed in all
the classical cadherins [33]. desmosome-bearing tissues and represent the predom-
Desmosomal cadherins are formed of five extracel- inant isoforms in simple epithelia [42,43], and are
lular cadherin repeats containing Ca2+ -binding sites mainly expressed in the basal layer of stratified epider-
and a cell-adhesion recognition (CAR) site [34,35]. All mis [32,41]. DSG4 is primarily expressed in the hair
DSC gene products undergo alternative splicing to gen- follicle and is restricted to the granular layer in strati-
erate a complete DSC ‘a’ form and a shortened DSC fied epithelia [44]. DSG1, DSG3 and DSG4, and DSC1
‘b’ form of the protein by the insertion of a mini-exon and DSG3, are predominantly expressed in the epider-
containing a stop codon, the length of their carboxy- mis, whereas DSG2 and DSC2 are highly expressed in
terminal domain being the only difference between the the myocardium [45]. Figure 2 summarizes the differ-
two isoforms [36]. ential expression patterns of desmosomal cadherins in
The desmosomal cadherins show complex develop- the epidermis.
mental and differentiation-specific patterns of expres- Within the cornified layer of the epidermis
sion [28], which implies that desmosomes within (stratum corneum), desmosomes are modified into
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 161

corneodesmosomes; structures that contain DSG1, that the transition from a Ca2+ -dependent to a Ca2+ -
DSC1 and corneodesmosin as their major extra- independent state with the induction of hyperadhesion
cellular constituents. The relative thickness of the proceeds via modulation of protein kinase C-α (PKCα)
stratum corneum is stabilized by the controlled degra- signalling.
dation of corneodesmosomes, any modifications at this Although the passage of desmosomes from a less
level leading to severe barrier defects [46]. Indeed, adhesive to a hyperadhesive state is Ca2+ -dependent,
mutations in the corneodesmosin (CDSN ) gene are O-glycosylation of the desmosomal plaque component
responsible for inherited skin and hair defects, such PG has also been shown to augment desmosomal
as peeling skin syndrome [47,48] and hypotrichosis adhesion in keratinocytes [61].
[49,50]. Meanwhile, impaired transport of cellular Whilst Ca2+ ions regulate the transition of desmo-
lipids and corneodesmosomal-associated proteases into somes from a hyperadhesive state into a less adhesive
the intercellular space of the epidermis, as a result of state and vice versa, there are other components, such
mutations in the transporter protein ABCA12, underlies as p120-catenin an armadillo-repeat protein, which
the pathogenesis of the severe and often lethal congen- regulate the formation of desmosomes. Kanno et al
ital disorder harlequin ichthyosis [51,52]. have observed that the interaction of p120-catenin with
DSGs and DSCs are required for strong cell–cell desmosomal cadherins such as DSG3 leads to desmo-
adhesion [53] via their interaction with each other some remodelling by maintaining free DSG3 at the
across the intercellular space, in a homophilic and/or cell surface before being internalized into desmosomes
heterophilic manner; the difference between the two [62].
types of interaction remaining unclear. The first types
of junction to form between adjacent cells are adherens
junctions, which create a first point of contact between Armadillo proteins
cells and facilitate the subsequent formation of desmo- PG (also called γ-catenin), together with PKPs1–3
somes in response to cell–cell contact, but also by [63,64] (all members of the armadillo family), are
raising extracellular levels of calcium (Ca2+ ) [54,55]. adaptor proteins with important roles in facilitating the
Via several binding motifs within their structure, DSGs adhesion of DSP to keratin intermediate filaments, in
and DSCs can bind Ca2+ and assume a rigidified regulating clustering of the desmosomal components
functional conformation [56], and thereby increase the and in mediating important signal transduction path-
level of adhesion between neighbouring cells and cre- ways. PG is formed of 12 arm repeats that share 65%
ate what has been described as ‘the dense midline of amino acid identity with β-catenin, the equivalent pro-
desmosomes’. In low-Ca2+ conditions the desmosomal tein associated with adherens junctions. The central
plaque components and membrane proteins are trans- armadillo domain of PG interacts with DSP, which
ported to the plasma membrane in separate compart- in turn tethers intermediate filaments to the desmo-
ments, but when desmosomal assembly is triggered, somal plaque. PG can also translocate to adherens
cadherin and DSP complexes do not associate as in nor- junctions and bind E-cadherin in the same manner as
mal Ca2+ conditions and remain separate [57]. It has β-catenin, but its higher affinity for DSP may explain
been observed that during the early stages of desmo- why PG and not β-catenin locates to desmosomes
some formation the assembly can reverse between [65].
the mature and young phases, but ultimately desmo- Although PKP1 and PKP2 can also localize to
somes mature and can no longer be dissociated by the nucleus, the PKPs are found predominantly in
calcium depletion [54]; the adhesion strength in cul- desmosomes [44]. PKPs1–3 share 50–55% sequence
tured keratinocytes increases after 6 days in culture similarity with the armadillo repeats of other armadillo
due to this phenomenon [57] in a similar way to family proteins [64]. Based on structural analysis
intact epithelia in vivo [58,59]. This is referred to as studies, PKPs contain nine arm repeat domains [66],
hyperadhesion and represents the result of high-affinity with 21 additional amino acids added to PKP1 and
and stable adhesive binding of desmosomal compo- 44 amino acids added to PKP2. Both PKP 1 and
nents into mature structures; it has not been observed PKP2 exist in two isoforms, a shorter ‘a’ form and
in adherens or tight junctions, making it specific to a longer ‘b’ form [67,68], with the short ‘a’ form more
desmosomes and explaining the hyperadhesive state of predominant.
keratinocytes [60]. PKPs show tissue- and differentiation-specific pat-
There are various situations in which desmosomes terns of expression similar to the desmosomal cad-
switch from a Ca2+ -independent to a Ca2+ -dependent herins. It has been observed that, while PKP3 shows
state. It has been observed that upon wounding of ker- expression throughout simple and all layers of strat-
atinocyte cell cultures, the desmosomes of cells situated ified epithelia, apart for hepatocytes, PKP1 is mostly
at the edge of the wound lose their hyperadhesiveness expressed in the suprabasal layers of stratified epithe-
and become Ca2+ -dependent [58], permitting the cell lia and PKP2 expression extends to simple epithelia,
motility required for wound re-epithelialization. Other lower layers of stratified epithelia and non-epithelial
situations include mitotically active basal cells and dur- tissues such as cardiac muscle—where it is the only
ing tumour invasion. Kimura et al [60] have shown isoform—and lymph nodes [67–72].
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
162 MA Brooke et al

PKPs appear to play a role in the clustering of both auto-immune and infectious diseases, which target
desmosomal proteins during the formation of desmo- the desmosomal cadherins.
somes. The N-terminal head domain of PKP1 can
associate with DSG1, PG, keratin and actin filaments Pemphigus vulgaris and pemphigus foliaceus
and ultimately with DSP through what appears to be
a robust association which drives DSP recruitment to The auto-immune diseases pemphigus vulgaris (PV)
cell–cell junctions [73–76]. PKP3 interacts with the and pemphigus foliaceus (PF) are a pair of potentially
largest number of desmosomal proteins, including DSP, fatal conditions characterized by loss of keratinocyte
PG, DSG1-3, DSC3a and 3b and DSC1a and 2a [64]. cell–cell adhesion (acantholysis) and blister forma-
PKP2 does play an important role in transport of DSP tion, in the epidermis (in PF) or mucosal membranes
to the plasma membrane during desmosome assem- (PV) [90]. Pemphigus auto-antibodies target the DSGs,
bly, but does so less efficiently than PKP1 [1,77]. with DSG3 identified as the auto-antigen in PV [91]
The mechanism behind PKP1 and PKP3 mediated- and DSG1 the auto-antigen in PF [92]. Among those
desmosomal assembly it is not yet fully determined, patients with early-stage PV, involving only mucous
although it appears that PKP2 functions as a scaffold membrane lesions, only anti-DSG3 auto-antibodies are
for PKC-α and regulates DSP association with inter- found, whilst those with later-stage disease, featuring
mediate filaments [1,78,79]. skin lesions in addition to mucosal, have sera con-
taining both anti-DSG1 and anti-DSG3 auto-antibodies
The plakin family [93].
The different patterns of disease in PV and PF
DSP, the most abundant desmosomal protein, plays a
reflect the distribution and expression levels of DSG1
key role as the linker between the plasma membrane
and DSG3, which differ significantly between the
and keratin intermediate filaments [44]. The protein
epidermis and mucosa [94]. In addition, the pattern
is predicted to form homodimers through a α-helical
of blister formation in the two conditions is influenced
coiled-coil rod domain which also interconnects a
by the apparent ability of DSG3 to compensate for
globular amino-terminus, responsible for binding the
adhesion defects caused by the presence of anti-
arm proteins PG and PKPs, and a carboxy-terminus
DSG1 antibodies, and vice versa—the so-called ‘DSG
domain, responsible for the attachment of intermediate
filaments [28,80–83]. Until recently only two isoforms compensation theory’ [94]. For example, blisters in
of DSP (DSPI and DSPII) have been known. As PF form only in the superficial epidermis because, as
with the ‘a’ and ‘b’ forms of desmocollins, DSPI and shown in Figure 2, this is the only area in which DSG1
DSPII isoforms are produced as a result of alternative is expressed in the absence of co-expressed DSG3.
mRNA splicing, with DSPII the shorter isoform of Meanwhile, the deeper blisters in later-stage PV reflect
the two. Both are widely expressed in numerous DSG3 expression in the absence of DSG1 in more basal
tissues, although DSPII expression is reduced/absent epidermal layers.
from the heart and from simple epithelia [84]. A The mechanism by which anti-DSG antibodies result
minor DSP isoform derived from DSPI, named DSPIα, in a loss of intracellular adhesion, and thus blister
produced by the alternative splicing of DSPI mRNA, formation, has been a matter of some controversy.
has recently been described and is detectable in lower Early research into the pathology of pemphigus auto-
levels than the dominant isoforms, although it presents antibodies investigated the potential of a role for
a similar tissue distribution [85]. proteases, particularly of the plasminogen activator
Franke et al have observed, by immunogold label- (PA) family, in indirectly mediating acantholysis and
ling of DSP, that in normal heart muscle DSP is blister formation [95,96] secondary to auto-antibody
located in all plaques of the desmosome-like and fascia binding. However, Mahoney et al [97] demonstrated
adherens-type junctions, with a very intense signal the pathogenicity of pemphigus auto-antibodies in a
within the desmosome-like junctions [86]. Several PA knock-out mouse model, demonstrating that these
studies in vivo and in vitro support the importance of proteases are not required for blister formation and
DSP in desmosome assembly and function, and show indicating that blister formation is a direct result of
that it appears to play a pivotal role in the development the effect of pemphigus auto-antibodies on DSGs.
of epidermis, neuroepithelium, heart and blood vessels The DSG3 epitopes recognized by auto-antibodies
[87,88]. In keratinocytes, DSPII appears to play a in PV patients have been found to reside particu-
more significant role than DSPI in maintaining robust larly in the DSG3 amino-terminal domain, the region
adhesion, suggesting cell type-specific functions for the implicated in forming the adhesive interface between
DSP isoforms [89]. neighbouring desmosomes [98,99], suggesting direct
steric interference in DSG ectodomain interactions as
the cause of acantholysis. However, keratinocytes have
Infectious and auto-immune diseases been shown to remain adhesive, with desmosomes
of desmosomes intact, when cells are incubated at 4 ◦ C with PV auto-
antibodies bound to DSG3, with several hours incuba-
The crucial role of desmosomes in mediating intracel- tion at 37 ◦ C instead required before significant loss of
lular adhesion is demonstrated by the consequences of adhesion is observed [100].
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 163

The consequent suggestion that a cellular response formation of blisters. The location and depth of the
by keratinocytes to PV auto-antibodies may be required blisters which result from ET action most likely again
for loss of adhesion is supported by observations of reflect the DSG compensation theory, as outlined for
DSG endocytosis, followed by desmosome disassem- PF, in that the blisters form in epidermal strata where
bly, in response to autoantibody binding in PV [1]. little to no DSG3 exists which could compensate for
PV auto-antibodies have been shown to cause rapid the loss of DSG1 function.
depletion of DSG3 from the membrane compartment Studies using time-lapse immunofluorescence and
of cultured cells [101]. This has been found to occur electron microscopy have shown that blister forma-
by endocytosis in sequential temporal phases, begin- tion in mice occurs following removal of the DSG1
ning with its internalization from non-desmosome- N-terminal domain, while the C-terminal remnant of
associated clusters at the cell surface (and delivered cleaved DSG1 remains at the cell surface [114]. This
to early endosomes) following initial binding of PV suggests that cleavage of DSG1 alone is sufficient
auto-antibodies [102]. This represents internalization to induce epidermal blister formation, rather than,
of newly-synthesized DSG3 not yet incorporated into for example, internalization of cleaved DSG1 being
desmosomes, and is also observed in studies utiliz- required. Work by Simpson et al [115] has further
ing anti-DSG3 monoclonal antibodies [103]. Indeed, demonstrated that 381-DSG1 remains bound to its
the pathogenicity of anti-DSG3 monoclonal antibodies catenin partner PG, effectively sequestering PG and
correlates with the extent to which they are capable of causing a dose-dependent reduction in other desmo-
depleting DSG3 from desmosomes [104]. This is fol- somal cadherins. This factor, along with the fact that
lowed by a re-arrangement of desmosomes into linear increased PG reduces cadherin expression, suggests
array structures, which serve as sites for endocytosis falling desmosomal cadherin levels as a result of PG
of desmosomal DSG3, resulting in junctional DSG3 sequestration by truncated DSG1 as a potential cellu-
depletion and loss of cell adhesion [105,106]. Signif- lar mechanism of acantholysis and blistering in bullous
icant internalization of desmosomal DSG3 does not impetigo and SSSS.
occur until 24 h after treatment of keratinocytes with
anti-DSG3 monoclonal antibodies [103], and occurs
in a clathrin- and dynamin-independent fashion [107]. Inherited desmosomal disorders
Thus, by inducing DSG3 internalization and thus
desmosome disassembly, PV auto-antibodies disrupt The complexity and relative lack of understanding
normal desmosomal homeostasis and trigger loss of of how desmosomal components interrelate with each
keratinocyte cell–cell adhesion and acantholysis [38]. other and with other compartments in a cell-type
and differentiation-dependent manner is reflected by
the range of genetic disorders arising from mutations
Bullous impetigo and staphylococcal scalded-skin
affecting the desmosomal genes. The large number of
syndrome
publications reporting various alterations affecting all
The infectious disease bullous impetigo, and its gen- of the desmosomal genes highlights the phenotypic
eralized form staphylococcal scalded skin syndrome heterogeneity behind these conditions; different muta-
(SSSS), conditions which mostly affect children under tions have been shown to result in either cardiac and/or
5 years of age and immunocompromised adults [108], cutaneous disorders, with or without hair implications.
are both characterized by epidermal blister formation One such condition frequently arising from desmoso-
as a result of keratinocyte acantholysis. Both of these mal mutations is arrhythmogenic right ventricular car-
related conditions share a startling clinical similarity diomyopathy (ARVC), an inherited disorder associated
to PF [109], with the histopathology of adult PF skin with arrhythmias and sudden cardiac death, and charac-
and that of neonatal mice with bullous impetigo being terized by fibro-fatty replacement of cardiac myocytes.
indistinguishable [110]. This is a result of the fact that ARVC-causing mutations have so far been identi-
the same desmosomal cadherin, DSG1, is affected in fied in a variety of desmosomal genes and account for
both cases, on this occasion by the exfoliative tox- 50–70% of ARVC cases. These include non-syndromic
ins (ETs), peptide toxins produced by some strains of ARVC mutations affecting all domains of DSP, which
pathogenic Staphylococcus aureus bacteria [110,111]. appear to be inherited in a dominant manner. More
The three known ETs affecting humans (ETA, ETB recently, a study of a German family has reported the
and ETD; murine and canine-specific forms also exist) first dominantly inherited ARVC-linked mutation in
are glutamate-specific serine proteases which specif- JUP (the gene encoding PG) [116,117]. All these vari-
ically cleave a single peptide bond in DSG1 (but ations cause ARVC without cutaneous abnormalities.
not the closely related DSG3), within its extracellu- Further, studies investigating the role of desmosomal
lar domain [112] and at a Ca2+ -binding site in which cadherins in human disease, particularly of the heart
the Ca2+ ion is required for cleavage to take place and skin, have revealed both autosomal dominant and
[113]. This removes residues 1–381 of the DSG1 recessive mutations in DSG2 and, more recently, a
ectodomain to produce the truncated protein 381- recessive mutation in DSC2, which also cause ARVC
DSG1. Direct DSG1 cleavage in this way serves to without a cutaneous or hair phenotype [118]. How-
disrupt keratinocyte cell–cell adhesion, leading to the ever, PKP2 is the gene most frequently associated with
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
164 MA Brooke et al

ARVC, with both dominant and recessive mutations in Homozygous mutations in the gene encoding PG
this gene identified as being responsible for up to 70% underlie an autosomal recessive condition known as
of cases. In the majority of cases PKP2 mutations were ‘Naxos disease’ (affected individuals hailing from
found in its carboxy-terminus domain, although other the Greek island of Naxos), which includes ARVC
mutations have also been reported [122]. (presenting without the pronounced left ventricular
Mutations in other PKPs appear to result in phe- involvement and early morbidity observed in Carvajal
notypes ranging from skin fragility to severe auto- syndrome) alongside woolly hair and PPK [133–135].
somal recessive ectodermal dysplasia, both due to a The epidermal manifestations in Naxos patients are not
variety of mutations in PKP1, which range from mis- as severe as those observed in PG-null mice [38]. Other
sense and nonsense mutations to splice-site and com- recent studies have described a recessive missense
pound heterozygous changes [119–122]. Experiments mutation affecting JUP in a patient who presented with
on PKP2-null mice have shown mid-gestational embry- ARVC, PPK and total alopecia [136], whilst Pigors
onic lethality caused by cardiac patterning defects and et al reported a novel lethal phenotype caused by a
fragility of the myocardium [123], alongside retraction nonsense mutation in JUP leading to severe congenital
of intermediate filaments from the plasma membrane, skin fragility with generalized epidermolysis, massive
demonstrating the importance of PKPs in DSP recruit- transcutaneous fluid loss and no apparent cardiac dys-
ment and intermediate filament tethering to desmo- function, due to a complete loss of PG in the patient’s
somes [44]. Although no disease-causing mutations skin leading to fewer desmosomes and no adhesion
have been reported in humans for PKP3, ablation of structures between keratinocytes [137]. Furthermore,
this isoform in mice results in defective hair folli- Cabral et al have described loss-of-function mutations
cle morphogenesis, increased keratinocyte proliferation (eg p.S24X) in the JUP gene, resulting in skin fragility,
and DSP mislocalization, leading to susceptibility to diffuse PPK and woolly hair without symptoms of
dermatitis and secondary alopecia [124]. cardiomyopathy [138]. Again, no PG expression was
DSP is the most abundant component within the detectable by immunostaining in skin sections from
desmosome and it is therefore not surprising that patients harbouring these mutations.
there are a variety of genetic disorders associated with With regard to other desmosomal cadherins, muta-
mutations in this gene, resulting in conditions with tions leading to loss of DSG4 are responsible for dis-
varying degrees of severity [125]. Previous studies ruptions in hair-follicle differentiation, whilst DSG1
have reported that DSP haploinsufficiency may cause haploinsufficiency leads to SPPK [139–141]. No
the epidermal-thickening disease striate palmoplantar disease-causing mutations in DSG3 have so far been
keratoderma (SPPK) [126], compound heterozygosity reported. As demonstrated by these examples, a most
with amino-terminal missense mutations, and carboxy- interesting aspect of inherited desmosomal disease is
terminal nonsense mutations leading to severe kera- the wide spectrum of conditions which can arise from
toderma, skin fragility and woolly hair, or alopecia mutations in some desmosomal components, such as
with or without cardiac involvement [117,127,128]. DSP and PG, in contrast to the restricted phenotypes
For example, a single base-pair deletion mutation in which result from mutations in the PKPs and desmo-
Ecuadorian families, that truncates the intermediate somal cadherins. This is illustrated in Figure 3.
filament-binding site of DSP, leads to the so-called
Carvajal syndrome, an ARVC variant which shows pre-
dominantly left ventricular involvement, early morbid- Other desmosome-related disorders
ity and clinical overlaps with dilated cardiomyopathy,
and is accompanied by woolly hair and palmoplan-
tar keratoderma (PPK) [128]. Another mutation which Darier’s disease
further truncates the carboxy-terminus of DSP is asso- The contribution of desmosomal regulation to disease
ciated with severe acantholytic epidermolysis bullosa, states is well illustrated by the example of Darier’s
a lethal disorder which presents complete alopecia, disease (also known as Darier–White disease or
neonatal teeth and nail loss and death due to transcu- dyskeratosis follicularis), an autosomal dominant skin
taneous fluid loss as a result of extensive skin erosion condition characterized by keratotic papules, which
[129]. form into larger skin lesions [142]. Histologically,
The critical role of the armadillo-family protein PG acantholysis (resulting from impaired desmosome for-
in desmosome assembly has also been demonstrated by mation and leading to suprabasal cleavage) is promi-
knock-out studies in mice, which show acantholysis, nent, accompanied by abnormal keratinization and
indicative of compromised desmosome function, and rounded keratinocytes [142]. Loss-of-function (hap-
are lethal due to fragility of the myocardium [130,131]. loinsufficiency) mutations in ATP2A2, encoding the
In some cases mouse pups were born but presented Ca2+ pump SERCA2, have been identified as the cause
epidermal fragility and heart defects and died shortly of this condition [143].
after birth. In another case, adult mice with a cardiac- As SERCA2 is responsible for Ca2+ transloca-
restricted deletion of PG presented with myocyte loss, tion from the cytosol to the endoplasmic reticulum
inflammation, fibrosis and cardiac dysfunction, and (ER) lumen [144], reduced SERCA2 function results
recapitulated the phenotype of ARVC [132]. in depleted ER Ca2+ stores; this can trigger an
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 165

Figure 3. Disease phenotypes resulting from mutations in selected desmosomal components. A wide spectrum of disease phenotypes can
result from mutations in either DSP or JUP (A), whilst mutations in the PKPs or desmosomal cadherins result in very limited disease spectra
(B). Mutations connected by lines represent reported compound heterozygous mutations. Note that several distinct mutations may be
represented by each symbol (eg several dominant mutations in the DSP N-terminus have been reported to cause ARVC alone). Adapted in
part from Bolling and Jonkman [116] and Cabral et al [131].

ER-stress response, leading to apoptosis in some ker- a recessive loss of function mutation in ADAM17
atinocytes [145,146] and also to impaired desmosome linked to a neonatal-onset inflammatory skin and bowel
formation, resulting to acantholysis. Studies using the disease [155], cutaneous features of which include
SERCA2 inhibitor thapsigargin (TG), have demon- a generalized pustular rash developing into psoriasi-
strated impaired trafficking of desmosomal compo- form erthyroderma, thickening of both the epidermis
nents, in particular DSP, and consequent impaired and nails, short and disorganized hair with abnormal-
desmosome formation in TG-treated cells compared to ities of the hair shaft (severe weathering and dam-
controls [147]. Meanwhile, cells that have undergone aged cuticles) and susceptibility to repeated bacterial
siRNA silencing of SERCA2 display significant reduc- infection. Immunofluorescence and western blot stud-
tions in translocation of DSP (reduced by 60%) and ies have revealed significant retention of DSG2 at the
PKCα (an important regulator of desmosomal assem- keratinocyte cell surface, suggesting that, in part, the
bly; reduced by 70%) to the plasma membrane [148]. skin and hair phenotypes observed may be mediated
by dysregulation of DSG cycling.
ADAM17 loss-of-function syndrome In addition, one of the patients identified with this
mutation died as a consequence of myocarditis caused
ADAM17 is a member of the a disintegrin and by Parvovirus B19 [155]. Considering the very well
metalloprotease (ADAM) family, consisting of established role for desmosomal components in ARVC,
membrane-anchored metalloproteases which mediate a link between desmosomal dysregulation and this
ectodomain shedding, the proteolytic release of extra- heart phenotype cannot be discounted. Furthermore,
cellular domains from membrane-bound precursors this may suggest a link between infectious myocardi-
[149]. First described as the tumour necrosis factor-α tis and desmosomal dysregulation in the absence of
converting enzyme [150,151], ADAM17 also cleaves ADAM17.
a wide variety of other proteins, including the desmo-
somal cadherin DSG2 [152], and has been shown to
cooperate with signalling through the epidermal growth Cystatin A and exfoliative ichthyosis
factor receptor (EGFR; a known regulator of desmo- Cystatin A (CSTA) belongs to the cystatin superfamily
somal assembly [153]) to regulate DSG2 shedding and of cysteine protease inhibitors and is expressed abun-
endocytic trafficking [154]. We have recently described dantly in the cytoplasm of epithelial and lymphoid
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
166 MA Brooke et al

tissues. CSTA is thought to play an important role in Studies on cultured cells have observed that, in some
many diverse mechanisms, from skin protection against cases, over-expression of desmosomal components pro-
allergens, such as dust mites [156], to regulating the motes proliferation, inhibits apoptosis and increases
activity of several target proteases in different types invasion, characteristics that are advantageous to
of cancers, including tumours of the breast [157], lung tumour cells; while conversely, other experiments have
[158], prostate [159] and squamous cell carcinomas of shown that over-expression of desmosomal compo-
the head and neck [160]. Some of the target proteases nents in cell lines suppresses such tumour-promoting
of CSTA, such as cathepsins B, H and L, are observed behaviour as invasion and anchorage-independent
to be frequently up-regulated in cancer, and appear growth [169].
to facilitate tumour invasion and metastasis through The desmosomal-related protein PERP (p53 apopto-
cleavage of cell–cell junctions [160]. sis effector related to PMP-22) has also been shown to
Recently, we have identified two unrelated families be associated with cancer. PERP is a membrane pro-
that presented homozygous loss-of-function mutations tein required for desmosome assembly (although its
in CSTA as being responsible for exfoliative ichthyosis, interacting partners remain incompletely understood),
a condition affecting the intercellular linkage in the and is also transcriptionally activated by the related
basal and suprabasal layers of the epidermis [161]. EM transcription factors p53 and p63 during DNA damage-
of basal and suprabasal layers of affected epidermis induced apoptosis and during the development of strat-
has shown increased intercellular gaps and thicken- ified epithelia, respectively. Studies have shown that
ing of keratin filaments. Experiments performed using mice with PERP knock-out or loss-of-function muta-
in vitro skin models mirrored that of patient skin, such tions develop a dramatic blistering phenotype in the
as breakage of intercellular connections upon stretch- epidermis and stratified epithelia [170], and also have
induced stress in CSTA knockdown monolayers and an increased tendency to develop squamous cell carci-
thicker keratin filaments in organotypic cultures lack- nomas when exposed to chronic ultraviolet light com-
ing CSTA. These observations reveal a previously pared to controls; with the loss of PERP in the skin
unknown role for CSTA in basal to suprabasal ker- reducing tumour latency and facilitating tumour pro-
atinocyte adhesion. gression over differentiation [171] [169]. A clear role
for PERP in mediating tumour suppression downstream
of the p53 and p63 transcription factors has since been
Non-adhering functions of desmosomal identified [169] [170], although its role in the desmo-
components in disease some remains unclear.

Desmosomal components in cancer Desmosomal components in signalling


It has been observed that the desmosome may also play and morphogenesis
an important role in cancer progression, with various In addition to their role in cell adhesion, desmoso-
components shown to be up-regulated, down-regulated mal components have various roles in signalling and
or modulated in a variety of tumour types. Attempts regulation in cells. Prominent examples of this feature
to clarify the role of desmosomal adhesion, and in are the desmosomal cadherins, whose distinct expres-
investigating the level of desmosomal involvement in sion patterns in epidermis (illustrated in Figure 2) have
cancers, have produced contradictory and confusing been shown to play a role in morphoregulation of
results. Studies have shown that an increased expres- that tissue. This can be mediated through either the
sion of desmosome proteins, such as DSG2, DSG3 adhesive function of cadherins [35] or the modula-
and PKP3, can be observed in certain cancers of the tion of intracellular signalling pathways, such as by
skin, head and neck, prostate and lung compared with inhibition of EGFR signalling [172] or via increas-
normal tissue, and that this over-expression is asso- ing β-catenin stability and signalling [173]. Further-
ciated with enhanced tumour progression [162–166]. more, studies in which various cadherins are forcibly
For example, two reports have shown that PG can expressed outside their usual strata in mouse epider-
transform cells by the specific activation of c-myc mis lead to disruption of normal epidermal structure
in a β-catenin-independent manner [167], whilst con- and function. Phenotypes such as epidermal hyperpro-
comitantly inhibiting apoptosis by induction of the liferation and abnormal differentiation [174,175], gross
anti-apoptotic protein Bcl-2 [168]. Meanwhile, in con- epidermal scaling [176], acantholysis and blistering
trast, the loss or reduction of one or more desmoso- (resembling PV) [177] and epidermal fragility along-
mal components, including DSG1-3, DSC2, DSC3, PG, side ulcerating lesions [178] have been observed in
PKP1-3 and DSP, has been observed in the develop- such studies, underlining the morphoregulatory impor-
ment or progression of various human epithelial can- tance of the desmosomal cadherins.
cers, such as skin, head and neck, gastric, colorectal, Among the other desmosomal components, the PKPs
bladder, breast, prostate, cervical and endometrial can- can function in the nucleus and may play a role in gene
cers [169]. Further, there are instances in which no regulation or nuclear structure [67], having been found
obvious changes in the level of desmosomal proteins to associate with proteins regulating rRNA and tRNA
have been observed during cancer progression [165]. transcription [179] and elF4A-dependent translation
Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 167

[180], among others. Meanwhile, PG can also function 15. Estivill X, Fortina P, Surrey S, et al . Connexin-26 mutations in
in both the desmosome and the nucleus, regulating Wnt sporadic and inherited sensorineural deafness. Lancet 1998; 351:
growth factor signalling alongside β-catenin [181,182] 394–398.
and regulating transcription of Tcf/Lef1 target genes, in 16. Marchiando AM, Graham WV, Turner JR. Epithelial barriers in
homeostasis and disease. Annu Rev Pathol 2010; 5: 119–144.
both a β-catenin-dependent and -independent fashion
17. Krause G, Winkler L, Mueller SL, et al . Structure and function
[183,184]. Indeed, suppression of Tcf–Lef1 signalling
of claudins. Biochim Biophys Acta 2008; 1778: 631–645.
may contribute to the pathology of ARVC, with sup- 18. Anderson JM, Van Itallie CM. Physiology and function of the
pression of DSP expression in mouse myocytes result- tight junction. Cold Spring Harb Perspect Biol 2009; 1: a002584.
ing in nuclear localization of PG, consequently causing 19. Schneeberger EE, Lynch RD. The tight junction: a multi-
suppression of Tcf–Lef1 signalling and enhanced car- functional complex. Am J Physiol Cell Physiol 2004; 286:
diac adipogenesis, fibrogenesis and myocyte apoptosis C1213–1228.
[185]. 20. Simon DB, Lu Y, Choate KA, et al . Paracellin-1, a renal tight
junction protein required for paracellular Mg2+ resorption. Sci-
Acknowledgment ence 1999; 285: 103–106.
21. Hou J, Paul DL, Goodenough DA. Paracellin-1 and the modula-
Funding was obtained from Barts and the London
tion of ion selectivity of tight junctions. J Cell Sci 2005; 118:
Charity. 5109–5118.
22. Konrad M, Schaller A, Seelow D, et al . Mutations in the tight-
junction gene claudin 19 (CLDN19 ) are associated with renal
Author contributions magnesium wasting, renal failure, and severe ocular involvement.
Am J Hum Genet 2006; 79: 949–957.
MAB, DN and DPK planned the manuscript structure 23. Carlton VE, Harris BZ, Puffenberger EG, et al . Complex inher-
and figures. The first draft was written by MAB and itance of familial hypercholanemia with associated mutations in
DN and revised by DPK. TJP2 and BAAT . Nat Genet 2003; 34: 91–96.
24. Riazuddin S, Ahmed ZM, Fanning AS, et al . Tricellulin is a
tight-junction protein necessary for hearing. Am J Hum Genet
References 2006; 79: 1040–1051.
25. Farquhar MG, Palade GE. Junctional complexes in various
1. Green KJ, Getsios S, Troyanovsky S, et al . Intercellular junction epithelia. J Cell Biol 1963; 17: 375–412.
assembly, dynamics, and homeostasis. Cold Spring Harb Perspect 26. Kelly DE. Fine structure of desmosomes, hemidesmosomes, and
Biol 2010; 2: a000125. an adepidermal globular layer in developing newt epidermis.
2. Meng W, Takeichi M. Adherens junction: molecular architecture J Cell Biol 1966; 28: 51–72.
and regulation. Cold Spring Harb Perspect Biol 2009; 1: a002899. 27. Staehelin LA. Structure and function of intercellular junctions. Int
3. Niessen CM, Gottardi CJ. Molecular components of the adherens Rev Cytol 1974; 39: 191–283.
junction. Biochim Biophys Acta 2008; 1778: 562–571. 28. Holthofer B, Windoffer R, Troyanovsky S, et al . Structure and
4. Yamada S, Pokutta S, Drees F, et al . Deconstructing the cad- function of desmosomes. Int Rev Cytol 2007; 264: 65–163.
herin–catenin–actin complex. Cell 2005; 123: 889–901. 29. Green KJ, Gaudry CA. Are desmosomes more than tethers for
5. Sprecher E, Bergman R, Richard G, et al . Hypotrichosis with
intermediate filaments? Nat Rev Mol Cell Biol 2000; 1: 208–216.
juvenile macular dystrophy is caused by a mutation in CDH3,
30. Odland GF. The fine structure of the interrelationship of cells in
encoding P-cadherin. Nat Genet 2001; 29: 134–136.
the human epidermis. J Biophys Biochem Cytol 1958; 4: 529–538.
6. Kjaer KW, Hansen L, Schwabe GC, et al . Distinct CDH3 muta-
31. Thomason HA, Scothern A, McHarg S, et al . Desmosomes:
tions cause ectodermal dysplasia, ectrodactyly, macular dystrophy
adhesive strength and signalling in health and disease. Biochem J
(EEM syndrome). J Med Genet 2005; 42: 292–298.
2010; 429: 419–433.
7. Shimomura Y, Wajid M, Shapiro L, et al . P-cadherin is a p63
32. Garrod DR, Merritt AJ, Nie Z. Desmosomal cadherins. Curr Opin
target gene with a crucial role in the developing human limb bud
Cell Biol 2002; 14: 537–545.
and hair follicle. Development 2008; 135: 743–753.
33. Kljuic A, Bauer RC, Christiano AM. Genomic organization of
8. Moon RT, Bowerman B, Boutros M, et al . The promise and
mouse desmocollin genes reveals evolutionary conservation. DNA
perils of Wnt signaling through β-catenin. Science 2002; 296:
Seq 2004; 15: 148–152.
1644–1646.
9. Polakis P. Wnt signaling and cancer. Genes Dev 2000; 14: 34. Tselepis C, Chidgey M, North A, et al . Desmosomal adhesion
1837–1851. inhibits invasive behavior. Proc Natl Acad Sci USA 1998; 95:
10. Ge X, Wang X. Role of Wnt canonical pathway in hematological 8064–8069.
malignancies. J Hematol Oncol 2010; 3: 33. 35. Runswick SK, O’Hare MJ, Jones L, et al . Desmosomal adhesion
11. Scott CA, Kelsell DP. Key functions for gap junctions in skin and regulates epithelial morphogenesis and cell positioning. Nat Cell
hearing. Biochem J 2011; 438: 245–254. Biol 2001; 3: 823–830.
12. Wei CJ, Xu X, Lo CW. Connexins and cell signaling in devel- 36. Collins JE, Legan PK, Kenny TP, et al . Cloning and sequence
opment and disease. Annu Rev Cell Dev Biol 2004; 20: analysis of desmosomal glycoproteins 2 and 3 (desmocollins):
811–838. cadherin-like desmosomal adhesion molecules with heteroge-
13. Kelsell DP, Di WL, Houseman MJ. Connexin mutations in skin neous cytoplasmic domains. J Cell Biol 1991; 113: 381–391.
disease and hearing loss. Am J Hum Genet 2001; 68: 37. Bannon LJ, Cabrera BL, Stack MS, et al . Isoform-specific dif-
559–568. ferences in the size of desmosomal cadherin/catenin complexes.
14. Kelsell DP, Dunlop J, Stevens HP, et al . Connexin 26 mutations J Invest Dermatol 2001; 117: 1302–1306.
in hereditary non-syndromic sensorineural deafness. Nature 1997; 38. Kottke MD, Delva E, Kowalczyk AP. The desmosome: cell sci-
387: 80–83. ence lessons from human diseases. J Cell Sci 2006; 119: 797–806.

Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
168 MA Brooke et al

39. King IA, Sullivan KH, Bennett R Jr, et al . The desmocollins wounding in cultured epithelial cells. Mol Biol Cell 2000; 11:
of human foreskin epidermis: identification and chromosomal 1077–1092.
assignment of a third gene and expression patterns of the three 59. Garrod DR, Berika MY, Bardsley WF, et al . Hyperadhesion in
isoforms. J Invest Dermatol 1995; 105: 314–321. desmosomes: its regulation in wound healing and possible rela-
40. Shimizu H, Masunaga T, Ishiko A, et al . Pemphigus vulgaris tionship to cadherin crystal structure. J Cell Sci 2005; 118:
and pemphigus foliaceus sera show an inversely graded binding 5743–5754.
pattern to extracellular regions of desmosomes in different layers 60. Kimura TE, Merritt AJ, Garrod DR. Calcium-independent
of human epidermis. J Invest Dermatol 1995; 105: 153–159. desmosomes of keratinocytes are hyperadhesive. J Invest
41. North AJ, Chidgey MA, Clarke JP, et al . Distinct desmocollin Dermatol 2007; 127: 775–781.
isoforms occur in the same desmosomes and show reciprocally 61. Hu P, Berkowitz P, Madden VJ, et al . Stabilization of
graded distributions in bovine nasal epidermis. Proc Natl Acad plakoglobin and enhanced keratinocyte cell–cell adhesion
Sci USA 1996; 93: 7701–7705. by intracellular O-glycosylation. J Biol Chem 2006; 281:
42. Legan PK, Yue KK, Chidgey MA, et al . The bovine desmocollin 12786–12791.
family: a new gene and expression patterns reflecting epithelial 62. Kanno M, Isa Y, Aoyama Y, et al . P120–catenin is a
cell proliferation and differentiation. J Cell Biol 1994; 126: novel desmoglein 3 interacting partner: identification of the
507–518. p120–catenin association site of desmoglein 3. Exp Cell Res
43. Schafer S, Stumpp S, Franke WW. Immunological identification 2008; 314: 1683–1692.
and characterization of the desmosomal cadherin Dsg2 in coupled 63. Hatzfeld M. The p120 family of cell adhesion molecules. Eur
and uncoupled epithelial cells and in human tissues. Differentia- J Cell Biol 2005; 84: 205–214.
tion 1996; 60: 99–108. 64. Hatzfeld M. Plakophilins: Multifunctional proteins or just regula-
44. Delva E, Tucker DK, Kowalczyk AP. The desmosome. Cold tors of desmosomal adhesion? Biochim Biophys Acta 2007; 1773:
Spring Harb Perspect Biol 2009; 1: a002543. 69–77.
45. Li J, Radice GL. A new perspective on intercalated disc organi- 65. Choi HJ, Gross JC, Pokutta S, et al . Interactions of plakoglobin
zation: implications for heart disease. Dermatol Res Pract 2010; and β-catenin with desmosomal cadherins: basis of selective
2010: 207835. exclusion of α- and β-catenin from desmosomes. J Biol Chem
46. Ishida-Yamamoto A, Igawa S, Kishibe M. Order and disorder in 2009; 284: 31776–31788.
corneocyte adhesion. J Dermatol 2011; 38: 645–654. 66. Choi HJ, Weis WI. Structure of the armadillo repeat domain of
47. Oji V, Eckl KM, Aufenvenne K, et al . Loss of corneodesmosin plakophilin 1. J Mol Biol 2005; 346: 367–376.
leads to severe skin barrier defect, pruritus, and atopy: unraveling 67. Mertens C, Kuhn C, Franke WW. Plakophilins 2a and 2b: con-
the peeling skin disease. Am J Hum Genet 2010; 87: 274–281. stitutive proteins of dual location in the karyoplasm and the
48. Israeli S, Zamir H, Sarig O, et al . Inflammatory peeling skin syn- desmosomal plaque. J Cell Biol 1996; 135: 1009–1025.
drome caused by a mutation in CDSN encoding corneodesmosin. 68. Schmidt A, Langbein L, Rode M, et al . Plakophilins 1a and 1b:
J Invest Dermatol 2011; 131: 779–781. widespread nuclear proteins recruited in specific epithelial cells
49. Levy-Nissenbaum E, Betz RC, Frydman M, et al . Hypotrichosis as desmosomal plaque components. Cell Tissue Res 1997; 290:
simplex of the scalp is associated with nonsense mutations in 481–499.
CDSN encoding corneodesmosin. Nat Genet 2003; 34: 151–153. 69. Heid HW, Schmidt A, Zimbelmann R, et al . Cell type-specific
50. Davalos NO, Garcia-Vargas A, Pforr J, et al . A non-sense muta- desmosomal plaque proteins of the plakoglobin family:
tion in the corneodesmosin gene in a Mexican family with plakophilin 1 (band 6 protein). Differentiation 1994; 58: 113–131.
hypotrichosis simplex of the scalp. Br J Dermatol 2005; 153: 70. Mertens C, Kuhn C, Moll R, et al . Desmosomal plakophilin 2
1216–1219. as a differentiation marker in normal and malignant tissues.
51. Kelsell DP, Norgett EE, Unsworth H, et al . Mutations in Differentiation 1999; 64: 277–290.
ABCA12 underlie the severe congenital skin disease harlequin 71. Franke WW, Schumacher H, Borrmann CM, et al . The area com-
ichthyosis. Am J Hum Genet 2005; 76: 794–803. posita of adhering junctions connecting heart muscle cells of
52. Thomas AC, Tattersall D, Norgett EE, et al . Premature terminal vertebrates—III: assembly and disintegration of intercalated disks
differentiation and a reduction in specific proteases associated in rat cardiomyocytes growing in culture. Eur J Cell Biol 2007;
with loss of ABCA12 in Harlequin ichthyosis. Am J Pathol 2009; 86: 127–142.
174: 970–978. 72. Bonne S, van Hengel J, Nollet F, et al . Plakophilin-3, a novel
53. Getsios S, Amargo EV, Dusek RL, et al . Coordinated expression armadillo-like protein present in nuclei and desmosomes of
of desmoglein 1 and desmocollin 1 regulates intercellular adhe- epithelial cells. J Cell Sci 1999; 112 (Pt 14): 2265–2276.
sion. Differentiation 2004; 72: 419–433. 73. Kowalczyk AP, Hatzfeld M, Bornslaeger EA, et al . The head
54. Watt FM, Mattey DL, Garrod DR. Calcium-induced reorganiza- domain of plakophilin-1 binds to desmoplakin and enhances its
tion of desmosomal components in cultured human keratinocytes. recruitment to desmosomes. Implications for cutaneous disease.
J Cell Biol 1984; 99: 2211–2215. J Biol Chem 1999; 274: 18145–18148.
55. Jones JC, Goldman RD. Intermediate filaments and the initiation 74. Hatzfeld M, Haffner C, Schulze K, et al . The function of
of desmosome assembly. J Cell Biol 1985; 101: 506–517. plakophilin 1 in desmosome assembly and actin filament orga-
56. Pokutta S, Weis WI. Structure and mechanism of cadherins and nization. J Cell Biol 2000; 149: 209–222.
catenins in cell–cell contacts. Annu Rev Cell Dev Biol 2007; 23: 75. Wahl JK, 3rd. A role for plakophilin-1 in the initiation of
237–261. desmosome assembly. J Cell Biochem 2005; 96: 390–403.
57. Cirillo N, Lanza A, Prime SS. Induction of hyper-adhesion atten- 76. Hofmann I, Mertens C, Brettel M, et al . Interaction of
uates autoimmune-induced keratinocyte cell–cell detachment and plakophilins with desmoplakin and intermediate filament
processing of adhesion molecules via mechanisms that involve proteins: an in vitro analysis. J Cell Sci 2000; 113(Pt 13):
PKC. Exp Cell Res 2010; 316: 580–592. 2471–2483.
58. Wallis S, Lloyd S, Wise I, et al . The α-isoform of protein kinase 77. Chen X, Bonne S, Hatzfeld M, et al . Protein binding and func-
C is involved in signaling the response of desmosomes to tional characterization of plakophilin 2. Evidence for its diverse

Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 169

roles in desmosomes and β-catenin signaling. J Biol Chem 2002; the prevention of pemphigus IgG-induced acantholysis by a puri-
277: 10512–10522. fied plasminogen activator inhibitor. J Invest Dermatol 1989; 92:
78. Godsel LM, Hsieh SN, Amargo EV, et al . Desmoplakin assem- 310–314.
bly dynamics in four dimensions: multiple phases differentially 97. Mahoney MG, Wang ZH, Stanley JR. Pemphigus vulgaris and
regulated by intermediate filaments and actin. J Cell Biol 2005; pemphigus foliaceus antibodies are pathogenic in plasminogen
171: 1045–1059. activator knockout mice. J Invest Dermatol 1999; 113: 22–25.
79. Bass-Zubek AE, Hobbs RP, Amargo EV, et al . Plakophilin 2: a 98. Tsunoda K, Ota T, Aoki M, et al . Induction of pemphigus phe-
critical scaffold for PKC-α that regulates intercellular junction notype by a mouse monoclonal antibody against the amino-
assembly. J Cell Biol 2008; 181: 605–613. terminal adhesive interface of desmoglein 3. J Immunol 2003;
80. Kowalczyk AP, Stappenbeck TS, Parry DA, et al . Structure 170: 2170–2178.
and function of desmosomal transmembrane core and plaque 99. Amagai M, Ishii K, Hashimoto T, et al . Conformational epitopes
molecules. Biophys Chem 1994; 50: 97–112. of pemphigus antigens (Dsg1 and Dsg3) are calcium dependent
81. Bornslaeger EA, Godsel LM, Corcoran CM, et al . Plakophilin and glycosylation independent. J Invest Dermatol 1995; 105:
1 interferes with plakoglobin binding to desmoplakin, yet 243–247.
together with plakoglobin promotes clustering of desmosomal 100. Calkins CC, Setzer SV, Jennings JM, et al . Desmoglein endocy-
plaque complexes at cell–cell borders. J Cell Sci 2001; 114: tosis and desmosome disassembly are coordinated responses to
727–738. pemphigus autoantibodies. J Biol Chem 2006; 281: 7623–7634.
82. Choi HJ, Park-Snyder S, Pascoe LT, et al . Structures of two 101. Aoyama Y, Kitajima Y. Pemphigus vulgaris-IgG causes a rapid
intermediate filament-binding fragments of desmoplakin reveal depletion of desmoglein 3 (Dsg3) from the Triton X-100 soluble
a unique repeat motif structure. Nat Struct Biol 2002; 9: pools, leading to the formation of Dsg3-depleted desmosomes in
612–620. a human squamous carcinoma cell line, DJM-1 cells. J Invest
83. Yin T, Green KJ. Regulation of desmosome assembly and adhe- Dermatol 1999; 112: 67–71.
sion. Semin Cell Dev Biol 2004; 15: 665–677. 102. Sato M, Aoyama Y, Kitajima Y. Assembly pathway of
84. Angst BD, Nilles LA, Green KJ. Desmoplakin II expression is desmoglein 3 to desmosomes and its perturbation by pem-
not restricted to stratified epithelia. J Cell Sci 1990; 97: 247–257.
phigus vulgaris-IgG in cultured keratinocytes, as revealed by
85. Cabral RM, Wan H, Cole CL, et al . Identification and character-
time-lapsed labeling immunoelectron microscopy. Lab Invest
ization of DSPIa, a novel isoform of human desmoplakin. Cell
2000; 80: 1583–1592.
Tissue Res 2010; 341: 121–129.
103. Mao X, Choi EJ, Payne AS. Disruption of desmosome assembly
86. Franke WW, Borrmann CM, Grund C, et al . The area composita
by monovalent human pemphigus vulgaris monoclonal antibodies.
of adhering junctions connecting heart muscle cells of vertebrates.
J Invest Dermatol 2009; 129: 908–918.
I. Molecular definition in intercalated disks of cardiomyocytes by
104. Yamamoto Y, Aoyama Y, Shu E, et al . Anti-desmoglein 3
immunoelectron microscopy of desmosomal proteins. Eur J Cell
(Dsg3) monoclonal antibodies deplete desmosomes of Dsg3 and
Biol 2006; 85: 69–82.
differ in their Dsg3-depleting activities related to pathogenicity.
87. Gallicano GI, Bauer C, Fuchs E. Rescuing desmoplakin function
J Biol Chem 2007; 282: 17866–17876.
in extra-embryonic ectoderm reveals the importance of this
105. Jennings JM, Tucker DK, Kottke MD, et al . Desmosome disas-
protein in embryonic heart, neuroepithelium, skin and vasculature.
sembly in response to pemphigus vulgaris IgG occurs in distinct
Development 2001; 128: 929–941.
phases and can be reversed by expression of exogenous Dsg3.
88. Vasioukhin V, Bowers E, Bauer C, et al . Desmoplakin is essen-
J Invest Dermatol 2011; 131: 706–718.
tial in epidermal sheet formation. Nat Cell Biol 2001; 3:
106. Aoyama Y, Nagai M, Kitajima Y. Binding of pemphigus vulgaris
1076–1085.
IgG to antigens in desmosome core domains excludes immune
89. Cabral RM, Tattersall D, Patel V, et al . The DSPII splice variant
complexes rather than directly splitting desmosomes. Br J Der-
is critical for desmosome-mediated HaCaT keratinocyte adhesion.
J Invest Dermatol 2011; 131: S30. matol 2010; 162: 1049–1055.
90. Payne AS, Hanakawa Y, Amagai M, et al . Desmosomes and 107. Delva E, Jennings JM, Calkins CC, et al . Pemphigus vulgaris
disease: pemphigus and bullous impetigo. Curr Opin Cell Biol IgG-induced desmoglein-3 endocytosis and desmosomal disas-
2004; 16: 536–543. sembly are mediated by a clathrin- and dynamin-independent
91. Amagai M, Klaus-Kovtun V, Stanley JR. Autoantibodies against mechanism. J Biol Chem 2008; 283: 18303–18313.
a novel epithelial cadherin in pemphigus vulgaris, a disease of 108. Nishifuji K, Sugai M, Amagai M. Staphylococcal exfoliative tox-
cell adhesion. Cell 1991; 67: 869–877. ins: ‘molecular scissors’ of bacteria that attack the cutaneous
92. Ishii K, Amagai M, Hall RP, et al . Characterization of autoan- defense barrier in mammals. J Dermatol Sci 2008; 49: 21–31.
tibodies in pemphigus using antigen-specific enzyme-linked 109. Stanley JR, Amagai M. Pemphigus, bullous impetigo, and the
immunosorbent assays with baculovirus-expressed recombinant staphylococcal scalded-skin syndrome. N Engl J Med 2006; 355:
desmogleins. J Immunol 1997; 159: 2010–2017. 1800–1810.
93. Ding X, Aoki V, Mascaro JM, Jr., et al . Mucosal and mucocu- 110. Amagai M, Matsuyoshi N, Wang ZH, et al . Toxin in bullous
taneous (generalized) pemphigus vulgaris show distinct autoanti- impetigo and staphylococcal scalded-skin syndrome targets
body profiles. J Invest Dermatol 1997; 109: 592–596. desmoglein 1. Nat Med 2000; 6: 1275–1277.
94. Shirakata Y, Amagai M, Hanakawa Y, et al . Lack of mucosal 111. Amagai M, Yamaguchi T, Hanakawa Y, et al . Staphylococcal
involvement in pemphigus foliaceus may be due to low expression exfoliative toxin B specifically cleaves desmoglein 1. J Invest
of desmoglein 1. J Invest Dermatol 1998; 110: 76–78. Dermatol 2002; 118: 845–850.
95. Hashimoto K, Shafran KM, Webber PS, et al . Anti-cell surface 112. Hanakawa Y, Schechter NM, Lin C, et al . Molecular mecha-
pemphigus autoantibody stimulates plasminogen activator activity nisms of blister formation in bullous impetigo and staphylococcal
of human epidermal cells. A mechanism for the loss of epidermal scalded skin syndrome. J Clin Invest 2002; 110: 53–60.
cohesion and blister formation. J Exp Med 1983; 157: 259–272. 113. Hanakawa Y, Selwood T, Woo D, et al . Calcium-dependent con-
96. Hashimoto K, Wun TC, Baird J, et al . Characterization of ker- formation of desmoglein 1 is required for its cleavage by exfo-
atinocyte plasminogen activator inhibitors and demonstration of liative toxin. J Invest Dermatol 2003; 121: 383–389.

Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
170 MA Brooke et al

114. Nishifuji K, Shimizu A, Ishiko A, et al . Removal of amino- 133. McKoy G, Protonotarios N, Crosby A, et al . Identification of
terminal extracellular domains of desmoglein 1 by staphylococcal a deletion in plakoglobin in arrhythmogenic right ventricular
exfoliative toxin is sufficient to initiate epidermal blister forma- cardiomyopathy with palmoplantar keratoderma and woolly hair
tion. J Dermatol Sci 2010; 59: 184–191. (Naxos disease). Lancet 2000; 355: 2119–2124.
115. Simpson CL, Kojima S, Cooper-Whitehair V, et al . Plakoglobin 134. Protonotarios N, Tsatsopoulou A. Naxos disease and Carvajal
rescues adhesive defects induced by ectodomain truncation of syndrome: cardiocutaneous disorders that highlight the pathogen-
the desmosomal cadherin desmoglein 1: implications for exfo- esis and broaden the spectrum of arrhythmogenic right ventricular
liative toxin-mediated skin blistering. Am J Pathol 2010; 177: cardiomyopathy. Cardiovasc Pathol 2004; 13: 185–194.
2921–2937. 135. Delmar M, McKenna WJ. The cardiac desmosome and arrhyth-
116. Bolling MC, Jonkman MF. Skin and heart: une liaison dan- mogenic cardiomyopathies: from gene to disease. Circ Res 2010;
gereuse. Exp Dermatol 2009; 18: 658–668. 107: 700–714.
117. Asimaki A, Syrris P, Wichter T, et al . A novel dominant muta- 136. Erken H, Yariz KO, Duman D, et al . Cardiomyopathy with
tion in plakoglobin causes arrhythmogenic right ventricular car- alopecia and palmoplantar keratoderma (CAPK) is caused by a
diomyopathy. Am J Hum Genet 2007; 81: 964–973. JUP mutation. Br J Dermatol 2011.
118. Simpson MA, Mansour S, Ahnood D, et al . Homozygous muta- 137. Pigors M, Kiritsi D, Krumpelmann S, et al . Lack of plakoglobin
tion of desmocollin-2 in arrhythmogenic right ventricular car- leads to lethal congenital epidermolysis bullosa: a novel clinico-
diomyopathy with mild palmoplantar keratoderma and woolly genetic entity. Hum Mol Genet 2011; 20: 1811–1819.
hair. Cardiology 2009; 113: 28–34. 138. Cabral RM, Liu L, Hogan C, et al . Homozygous mutations in
119. Pieperhoff S, Barth M, Rickelt S, et al . Desmosomal molecules the 5 region of the JUP gene result in cutaneous disease but
in and out of adhering junctions: normal and diseased states of normal heart development in children. J Invest Dermatol 2010;
epidermal, cardiac and mesenchymally derived cells. Dermatol 130: 1543–1550.
Res Pract 2010; 2010: 139167. 139. Kljuic A, Gilead L, Martinez-Mir A, et al . A nonsense mutation
120. Tanaka A, Lai-Cheong JE, Cafe ME, et al . Novel truncating in the desmoglein 1 gene underlies striate keratoderma. Exp
mutations in PKP1 and DSP cause similar skin phenotypes in
Dermatol 2003; 12: 523–527.
two Brazilian families. Br J Dermatol 2009; 160: 692–697.
140. Rickman L, Simrak D, Stevens HP, et al . N-terminal deletion
121. Zheng R, Bu DF, Zhu XJ. Compound heterozygosity for new
in a desmosomal cadherin causes the autosomal dominant skin
splice site mutations in the plakophilin 1 gene (PKP1 ) in a
disease striate palmoplantar keratoderma. Hum Mol Genet 1999;
Chinese case of ectodermal dysplasia–skin fragility syndrome.
8: 971–976.
Acta Derm Venereol 2005; 85: 394–399.
141. Awad MM, Dalal D, Cho E, et al . DSG2 mutations contribute to
122. Ersoy-Evans S, Erkin G, Fassihi H, et al . Ectodermal dyspla-
arrhythmogenic right ventricular dysplasia/cardiomyopathy. Am
sia–skin fragility syndrome resulting from a new homozygous
J Hum Genet 2006; 79: 136–142.
mutation, 888delC, in the desmosomal protein plakophilin 1. J Am
142. Savignac M, Edir A, Simon M, et al . Darier disease : a disease
Acad Dermatol 2006; 55: 157–161.
model of impaired calcium homeostasis in the skin. Biochim
123. Grossmann KS, Grund C, Huelsken J, et al . Requirement of
Biophys Acta 2011; 1813: 1111–1117.
plakophilin 2 for heart morphogenesis and cardiac junction
143. Sakuntabhai A, Ruiz-Perez V, Carter S, et al . Mutations in
formation. J Cell Biol 2004; 167: 149–160.
ATP2A2, encoding a Ca2+ pump, cause Darier disease. Nat Genet
124. Sklyarova T, Bonne S, D’Hooge P, et al . Plakophilin-3-deficient
1999; 21: 271–277.
mice develop hair coat abnormalities and are prone to cutaneous
144. Hovnanian A. SERCA pumps and human diseases. Subcell
inflammation. J Invest Dermatol 2008; 128: 1375–1385.
Biochem 2007; 45: 337–363.
125. Lai Cheong JE, Wessagowit V, McGrath JA. Molecular abnor-
145. Yoshida H. ER stress and diseases. FEBS J 2007; 274: 630–658.
malities of the desmosomal protein desmoplakin in human dis-
146. Pasmatzi E, Badavanis G, Monastirli A, et al . Reduced expres-
ease. Clin Exp Dermatol 2005; 30: 261–266.
126. Armstrong DK, McKenna KE, Purkis PE, et al . Haploinsuffi- sion of the antiapoptotic proteins of Bcl-2 gene family in the
ciency of desmoplakin causes a striate subtype of palmoplantar lesional epidermis of patients with Darier’s disease. J Cutan
keratoderma. Hum Mol Genet 1999; 8: 143–148. Pathol 2007; 34: 234–238.
127. Whittock NV, Wan H, Morley SM, et al . Compound heterozy- 147. Stuart RO, Sun A, Bush KT, et al . Dependence of epithelial
gosity for non-sense and mis-sense mutations in desmoplakin intercellular junction biogenesis on thapsigargin-sensitive intra-
underlies skin fragility/woolly hair syndrome. J Invest Dermatol cellular calcium stores. J Biol Chem 1996; 271: 13636–13641.
2002; 118: 232–238. 148. Hobbs RP, Amargo EV, Somasundaram A, et al . The calcium
128. Norgett EE, Hatsell SJ, Carvajal-Huerta L, et al . Recessive muta- ATPase SERCA2 regulates desmoplakin dynamics and intercellu-
tion in desmoplakin disrupts desmoplakin-intermediate filament lar adhesive strength through modulation of PKC and α-signaling.
interactions and causes dilated cardiomyopathy, woolly hair and FASEB J 2011; 25: 990–1001.
keratoderma. Hum Mol Genet 2000; 9: 2761–2766. 149. White JM. ADAMs: modulators of cell–cell and cell–matrix
129. Jonkman MF, Pasmooij AM, Pasmans SG, et al . Loss of desmo- interactions. Curr Opin Cell Biol 2003; 15: 598–606.
plakin tail causes lethal acantholytic epidermolysis bullosa. Am 150. Black RA, Rauch CT, Kozlosky CJ, et al . A metalloproteinase
J Hum Genet 2005; 77: 653–660. disintegrin that releases tumour-necrosis factor-α from cells.
130. Bierkamp C, McLaughlin KJ, Schwarz H, et al . Embryonic heart Nature 1997; 385: 729–733.
and skin defects in mice lacking plakoglobin. Dev Biol 1996; 180: 151. Moss ML, Jin SL, Milla ME, et al . Cloning of a disintegrin
780–785. metalloproteinase that processes precursor tumour-necrosis factor-
131. Acehan D, Petzold C, Gumper I, et al . Plakoglobin is required for α. Nature 1997; 385: 733–736.
effective intermediate filament anchorage to desmosomes. J Invest 152. Bech-Serra JJ, Santiago-Josefat B, Esselens C, et al . Proteomic
Dermatol 2008; 128: 2665–2675. identification of desmoglein 2 and activated leukocyte cell
132. Li J, Swope D, Raess N, et al . Cardiac tissue-restricted deletion adhesion molecule as substrates of ADAM17 and ADAM10
of plakoglobin results in progressive cardiomyopathy and activa- by difference gel electrophoresis. Mol Cell Biol 2006; 26:
tion of β-catenin signaling. Mol Cell Biol 2011; 31: 1134–1144. 5086–5095.

Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Cell–cell connectivity: desmosomes and disease 171

153. Lorch JH, Klessner J, Park JK, et al . Epidermal growth factor 169. Dusek RL, Attardi LD. Desmosomes: new perpetrators in tumour
receptor inhibition promotes desmosome assembly and strength- suppression. Nat Rev Cancer 2011; 11: 317–323.
ens intercellular adhesion in squamous cell carcinoma cells. J Biol 170. Ihrie RA, Marques MR, Nguyen BT, et al . Perp is a p63-
Chem 2004; 279: 37191–37200. regulated gene essential for epithelial integrity. Cell 2005; 120:
154. Klessner JL, Desai BV, Amargo EV, et al . EGFR and ADAMs 843–856.
cooperate to regulate shedding and endocytic trafficking of the 171. Beaudry VG, Jiang D, Dusek RL, et al . Loss of the p53/p63
desmosomal cadherin desmoglein 2. Mol Biol Cell 2009; 20: regulated desmosomal protein Perp promotes tumorigenesis. PLoS
328–337. Genet 2010; 6: e1001168.
155. Blaydon DC, Biancheri P, Di W-L, et al . Inflammatory skin and 172. Getsios S, Simpson CL, Kojima S, et al . Desmoglein 1-
bowel disease linked to deletion in ADAM17. N Engl J Med 2011; dependent suppression of EGFR signaling promotes epider-
365: 1502–1508. mal differentiation and morphogenesis. J Cell Biol 2009; 185:
156. Kato T, Takai T, Mitsuishi K, et al . Cystatin A inhibits IL-8 1243–1258.
production by keratinocytes stimulated with Der p 1 and Der f 1: 173. Hardman MJ, Liu K, Avilion AA, et al . Desmosomal cadherin
biochemical skin barrier against mite cysteine proteases. J Allergy misexpression alters β-catenin stability and epidermal differenti-
Clin Immunol 2005; 116: 169–176. ation. Mol Cell Biol 2005; 25: 969–978.
157. Kuopio T, Kankaanranta A, Jalava P, et al . Cysteine proteinase 174. Merritt AJ, Berika MY, Zhai W, et al . Suprabasal desmoglein
inhibitor cystatin A in breast cancer. Cancer Res 1998; 58: 3 expression in the epidermis of transgenic mice results in
432–436. hyperproliferation and abnormal differentiation. Mol Cell Biol
158. Butler MW, Fukui T, Salit J, et al . Modulation of cystatin A 2002; 22: 5846–5858.
expression in human airway epithelium related to genotype, smok- 175. Brennan D, Hu Y, Joubeh S, et al . Suprabasal Dsg2 expression in
ing, COPD, and lung cancer. Cancer Res 2011; 71: 2572–2581. transgenic mouse skin confers a hyperproliferative and apoptosis-
159. Sinha AA, Quast BJ, Korkowski JC, et al . The relationship of resistant phenotype to keratinocytes. J Cell Sci 2007; 120:
cathepsin B and stefin A mRNA localization identifies a potentially 758–771.
aggressive variant of human prostate cancer within a Gleason 176. Elias PM, Matsuyoshi N, Wu H, et al . Desmoglein isoform dis-
histologic score. Anticancer Res 1999; 19: 2821–2829. tribution affects stratum corneum structure and function. J Cell
160. Strojan P, Budihna M, Smid L, et al . Prognostic significance of Biol 2001; 153: 243–249.
cysteine proteinases cathepsins B and L and their endogenous 177. Chen J, Den Z, Koch PJ. Loss of desmocollin 3 in mice leads to
inhibitors stefins A and B in patients with squamous cell epidermal blistering. J Cell Sci 2008; 121: 2844–2849.
carcinoma of the head and neck. Clin Cancer Res 2000; 6: 178. Chidgey M, Brakebusch C, Gustafsson E, et al . Mice lacking
1052–1062. desmocollin 1 show epidermal fragility accompanied by barrier
161. Blaydon DC, Nitoiu D, Eckl KM, et al . Mutations in cystatin A defects and abnormal differentiation. J Cell Biol 2001; 155:
underlie autosomal recessive exfoliative ichthyosis and reveal a 821–832.
role for this protease inhibitor in cell–cell adhesion. Am J Hum 179. Mertens C, Hofmann I, Wang Z, et al . Nuclear particles contain-
Genet 2011; 89: 1–8. ing RNA polymerase III complexes associated with the junctional
162. Brennan D, Mahoney MG. Increased expression of Dsg2 in plaque protein plakophilin 2. Proc Natl Acad Sci USA 2001; 98:
malignant skin carcinomas: a tissue-microarray based study. Cell 7795–7800.
Adh Migr 2009; 3: 148–154. 180. Wolf A, Krause-Gruszczynska M, Birkenmeier O, et al .
163. Chen YJ, Chang JT, Lee L, et al . DSG3 is overexpressed in head Plakophilin 1 stimulates translation by promoting eIF4A1
and neck cancer and is a potential molecular target for inhibition activity. J Cell Biol 2010; 188: 463–471.
of oncogenesis. Oncogene 2007; 26: 467–476. 181. Huelsken J, Behrens J. The Wnt signalling pathway. J Cell Sci
164. Furukawa C, Daigo Y, Ishikawa N, et al . Plakophilin 3 oncogene 2002; 115: 3977–3978.
as prognostic marker and therapeutic target for lung cancer. 182. Chien AJ, Conrad WH, Moon RT. A Wnt survival guide: from
Cancer Res 2005; 65: 7102–7110. flies to human disease. J Invest Dermatol 2009; 129: 1614–1627.
165. Kurzen H, Munzing I, Hartschuh W. Expression of desmosomal 183. Zhurinsky J, Shtutman M, Ben-Ze’ev A. Plakoglobin and β-
proteins in squamous cell carcinomas of the skin. J Cutan Pathol catenin: protein interactions, regulation and biological roles.
2003; 30: 621–630. J Cell Sci 2000; 113(Pt 18): 3127–3139.
166. Breuninger S, Reidenbach S, Sauer CG, et al . Desmosomal 184. Conacci-Sorrell ME, Ben-Yedidia T, Shtutman M, et al . Nr-
plakophilins in the prostate and prostatic adenocarcinomas: impli- CAM is a target gene of the β-catenin/LEF-1 pathway in
cations for diagnosis and tumor progression. Am J Pathol 2010; melanoma and colon cancer and its expression enhances motility
176: 2509–2519. and confers tumorigenesis. Genes Dev 2002; 16: 2058–2072.
167. Kolligs FT, Kolligs B, Hajra KM, et al . γ-Catenin is regulated by 185. Garcia-Gras E, Lombardi R, Giocondo MJ, et al . Suppression of
the APC tumor suppressor and its oncogenic activity is distinct canonical Wnt/β-catenin signaling by nuclear plakoglobin recapit-
from that of β-catenin. Genes Dev 2000; 14: 1319–1331. ulates phenotype of arrhythmogenic right ventricular cardiomy-
168. Hakimelahi S, Parker HR, Gilchrist AJ, et al . Plakoglobin regu- opathy. J Clin Invest 2006; 116: 2012–2021.
lates the expression of the anti-apoptotic protein BCL-2. J Biol
Chem 2000; 275: 10905–10911.

Copyright  2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 158–171
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

You might also like