Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics - Cold Spring Harb ...

Page created by Brett Barnes
 
CONTINUE READING
Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics - Cold Spring Harb ...
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 Desmosomes and Intermediate Filaments:
                 Their Consequences for Tissue Mechanics

                 Mechthild Hatzfeld,1 René Keil,1 and Thomas M. Magin2
                 1
                  Institute of Molecular Medicine, Division of Pathobiochemistry, Martin-Luther-University Halle-Wittenberg,
                  06114 Halle, Germany
                 2
                  Institute of Biology, Division of Cell and Developmental Biology and Saxonian Incubator for Clinical
                  Translation (SIKT), University of Leipzig, 04103 Leipzig, Germany
                 Correspondence: thomas.magin@uni-leipzig.de; mechthild.hatzfeld@medizin.uni-halle.de

                 Adherens junctions (AJs) and desmosomes connect the actin and keratin filament networks of
                 adjacent cells into a mechanical unit. Whereas AJs function in mechanosensing and in
                 transducing mechanical forces between the plasma membrane and the actomyosin cyto-
                 skeleton, desmosomes and intermediate filaments (IFs) provide mechanical stability required
                 to maintain tissue architecture and integrity when the tissues are exposed to mechanical
                 stress. Desmosomes are essential for stable intercellular cohesion, whereas keratins deter-
                 mine cell mechanics but are not involved in generating tension. Here, we summarize the
                 current knowledge of the role of IFs and desmosomes in tissue mechanics and discuss
                 whether the desmosome–keratin scaffold might be actively involved in mechanosensing
                 and in the conversion of chemical signals into mechanical strength.

                     he majority of tissues are constantly exposed                   epithelia that line organ and body surfaces to
                 T   to external forces, such as mechanical load,
                 stretch, and shear stress, in addition to intrinsic
                                                                                     provide structural support and serve as barriers
                                                                                     against diverse external stressors such as me-
                 forces generated by contractile elements inside                     chanical force, pathogens, toxins, and dehydra-
                 tissues. Both extrinsic and intrinsic forces con-                   tion. Epithelia contain two types of intercellular
                 tribute to tissue morphogenesis, homeostasis,                       adhesion complexes: adherens junctions (AJs)
                 and regeneration and affect cell shape, prolifer-                   and desmosomes, connected to the actin and
                 ation, and migration (Evans et al. 2013). Sensing                   keratin cytoskeleton, respectively (Fletcher and
                 and transmitting forces depend to a large extent                    Mullins 2010).
                 on tight interactions between cell adhesion                              The detailed molecular mechanisms that
                 complexes and the cytoskeleton. Mechanosens-                        underlie mechanotransduction are complex
                 ing and mechanotransduction can be defined as                       and only partially understood. Recent data in-
                 cellular processes that convert mechanical cues                     dicate that mechanosensor proteins can under-
                 into intracellular signaling (Furuse et al. 2002;                   go force-induced conformational changes that,
                 Huveneers and de Rooij 2013; Janmey et al.                          in turn, induce changes in their activity or affin-
                 2013). These processes are exemplified by                           ity for binding partners (Yonemura et al. 2010;

                 Editors: Carien M. Niessen and Alpha S. Yap
                 Additional Perspectives on Cell –Cell Junctions available at www.cshperspectives.org
                 Copyright # 2017 Cold Spring Harbor Laboratory Press; all rights reserved
                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157

                                                                                                                                      1
Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics - Cold Spring Harb ...
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                 Huveneers and de Rooij 2013). This can finally                         referred to recent reviews (Meens et al. 2013;
                 lead to the activation of chemical signaling                           Patel and Green 2014).
                 cascades. As discussed in Yap (2017), AJs func-
                 tion as mechanosensors (Huveneers and de
                 Rooij 2013; Yao et al. 2014; Ladoux et al. 2015;                       FUNCTION OF DESMOSOMES IN
                 Muhamed et al. 2016), whereas a role in force                          CONFERRING MECHANICAL STABILITY
                 sensing has so far not been attributed to desmo-
                                                                                        Composition and Structure of Desmosomes
                 somes. At the same time, desmosome-mediated
                 intercellular adhesion is much stronger than AJ-                       Desmosomes are intercellular junctions essen-
                 mediated cohesion as shown by the epithelial                           tial for mediating strong intercellular cohesion
                 sheet assay: Whereas depletion of the desmoso-                         (Garrod 2010; Green et al. 2010; Kowalczyk and
                 mal plaque component plakophilin 1 (PKP1) in                           Green 2013). They are composed of three pro-
                 keratinocytes disrupts epithelial cohesion on                          tein families. The desmosomal cadherins, des-
                 application of mechanical stress, knockdown                            mogleins (DSGs), and desmocollins (DSCs),
                 of the corresponding components from AJs,                              are transmembrane proteins whose extracellular
                 p120, or p0071/PKP4, has no immediate effect                           domains form the adhesive interface of the des-
                 on intercellular cohesion (Fig. 1). Thus, this                         mosome, whereas their cytoplasmic tails anchor
                 suggests that in tissues in which both junctions                       the armadillo proteins, plakoglobin (PG/JUP),
                 are present, AJs are important in mechanosens-                         and plakophilins 1 – 3 (PKPs) to the desmo-
                 ing, whereas desmosomes are crucial for provid-                        somal plaque. The armadillo proteins, in turn,
                 ing mechanical stability under force.                                  bind to desmoplakin (DSP), a member of the
                     Here, we will review the contribution of the                       plakin family of cytoskeleton-associated pro-
                 desmosome – keratin complex to mechanical                              teins. DSP links the desmosome to the keratin
                 integrity of epithelial barriers, in particular of                     filament network, which is essential to provide
                 the epidermis, and discuss their potential func-                       tensile strength (Fig. 2). The importance of des-
                 tion in sensing and transmission of forces. For                        mosomes for tissue integrity is highlighted by
                 the role of intercellular contacts and intermedi-                      the severe skin and cardiac defects that arise in
                 ate filaments (IFs) of the heart, the reader is                        autoimmune and genetic diseases.

                     A          Control-si    p0071-si        p120-si        PKP1-si        PKP3-si         B
                                                                                                                Control-si

                                                                                                                             p0071-si

                                                                                                                                                  PKP1-si

                                                                                                                                                            PKP3-si
                                                                                                                                        p120-si
                     rotation
                      Before

                                                                                                                                                                      p0071

                                                                                                                                                                      p120
                     rotation

                                                                                                                                                                      PKP1
                      5 min

                                                                                                                                                                      PKP3
                                                                                                                                                                      α-Tubulin
                     rotation
                      20 min

                 Figure 1. Dispase-based dissociation assay highlights the importance of desmosomes for intercellular cohesion.
                 Only the knockdown of the desmosomal plaque protein plakophilin 1 (PKP1) severely disturbed intercellular
                 cohesion of mouse keratinocytes grown for 24 h in a medium containing 1.2 mM Ca2þ. The knockdown of the
                 corresponding proteins from adherens junctions (AJs), p120, or p0071/PKP4 did not interfere with mechanical
                 resistance of mouse keratinocytes (A), although the respective protein amounts were considerably decreased as
                 shown by western blot (B).

                 2                       Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                                                                                      Desmosomes and Keratins in Tissue Mechanics

                                                     The desmosome–keratin scaffold in cell mechanics

                                                                           C

                   B

                                                                           A

                 Figure 2. The desmosome –keratin complex as a micromechanical scaffold during epidermal differentiation.
                 (A) Expression of keratins K5/14 and interaction with desmosomal protein isoforms forms stable cohesion
                 among cells and protects basal keratinocytes against mechanical stress. Under conditions of tissue homeostasis,
                 stable desmosome –keratin scaffolds prevail. (B) Cells react to wounding (activated keratinocytes) by
                 modulating their micromechanical properties through altering adhesion and the cytoskeleton. Underlying
                 mechanisms involve altered expression of isotype proteins and posttranslational modifications that can dimin-
                 ish adhesion and render cells more migratory. Expression of K6/16/17 coincides with dynamic desmosomes.
                 (C ) To withstand increased mechanical stress, for example, in upper strata of the epidermis, number and size of
                 desmosomes are increased and keratin filaments become more abundant, bundled by associated proteins and
                 possibly elevated interkeratin Cys crosslinks. Gray arrow in (C) indicates tissue differentiation. Desmosomes are
                 depicted to indicate stable adhesion (red), to demarcate dynamic, less adhesive (light red), and hyperadhesive
                 complexes (dark red). Keratin filaments are drawn to indicate stable networks (straight blue lines), dynamic,
                 less stable networks (dashed green lines) or to indicate highly resilient, bundled networks (orange thick lines).
                 The above properties result from expression of distinct isotypes, relative abundance, and posttranslational
                 modifications.

                      Desmosomal cadherins are divided into                         whereas DSG2, -3, and DSC2, -3 occur primar-
                 four DSGs and three DSCs. The DSG – DSC het-                       ily in the lower layers of the epidermis (Green
                 erodimers represent the basic adhesive unit of                     and Gaudry 2000).
                 desmosomes (Harrison et al. 2016). All DSGs                             Despite their critical roles in maintaining
                 form adhesive dimers with all DSC isoforms                         epidermal adhesion and integrity, desmosomes
                 with affinities characteristic of each DSG – DSC                   are highly dynamic entities and undergo con-
                 pair. In contrast, homophilic DSG – DSG and                        stant remodeling to allow for plasticity and cell
                 DSC – DSC trans-interactions were suppressed                       migration within the epidermis during epider-
                 by charged amino acids. In multilayered epithe-                    mal differentiation and regeneration (Green
                 lia such as the epidermis, desmosomal cadher-                      and Gaudry 2000; Garrod 2010; Kowalczyk
                 ins reveal differentiation-dependent expression.                   and Green 2013). Desmosome adhesion can
                 DSG1, -4, and DSC1 are expressed in flattened                      be regulated at several levels: Their composi-
                 cells of the upper granular and cornified layers,                  tion, as well as size and number, vary among

                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157     3
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                 tissues and among the individual layers of the                  somes (Gallicano et al. 1998). In an epidermis-
                 epidermis, and are controlled at the transcrip-                 specific DSP KO, intercellular separations were
                 tional, posttranscriptional, and posttransla-                   observed as expected (Vasioukhin et al. 2001).
                 tional levels.                                                  Surprisingly, desmosome number was unal-
                                                                                 tered although the lack of keratin association
                                                                                 compromised their function. PKPs were previ-
                 Evidence from Mouse Models and Human
                                                                                 ously considered as nonessential plaque pro-
                 Diseases
                                                                                 teins. However, PKP2 KO mice died around
                 Gene ablation in mice has established the con-                  day 11.5 of embryonic development because of
                 tribution of individual desmosomal proteins                     heart defects, indicating that at least one PKP
                 to mechanical stability and tissue integrity in                 is required for stable intercellular adhesion
                 vivo. These studies show (a) that many of the                   (Grossmann et al. 2004). In contrast, PKP32/2
                 desmosomal proteins are required for organis-                   mice were viable but developed hair abnormal-
                 mal survival and (b) that isotypes of DSGs,                     ities and increased inflammation of the skin,
                 DSCs, and PKPs have distinct functions in                       manifest in mice kept in a nonpathogen-free
                 vivo. For example, phenotypes observed after                    environment (Sklyarova et al. 2008). We have
                 ablation of the three DSC genes differ dramati-                 recently shown that PKP1 is essential for epider-
                 cally in their severity: Whereas a DSC2 knock-                  mal integrity in vivo. PKP1 KO mice died
                 out (KO) did not result in any obvious pheno-                   shortly after birth with defects in epidermal co-
                 type (Rimpler 2014), ablation of DSC1 led to                    hesion and barrier formation (Rietscher et al.
                 epidermal fragility with hyperproliferation and                 2016). Desmosomes were small and sparse in
                 dermatitis, but mice were viable and fertile                    the skin, as well as in cultured keratinocytes de-
                 (Chidgey et al. 2001). In contrast, DSC3 abla-                  rived from these mice, and cell separation oc-
                 tion resulted in preimplantation lethality, sug-                curred in the granular layers.
                 gesting a desmosome-independent role during                          Additional support for the importance of
                 early development (Den et al. 2006). Similarly,                 desmosomal adhesion in tissue integrity comes
                 ablation of DSG isotypes revealed distinct func-                from monogenetic human diseases. Ectoder-
                 tions in vivo: Whereas the DSG2 KO was em-                      mal dysplasia-skin fragility syndrome (EDSFS,
                 bryonic lethal (Eshkind et al. 2002), DSG3 KO                   MIM604536), a genetic disease caused by PKP1
                 mice showed weakened desmosomal adhesion,                       mutations (McGrath et al. 1997), is character-
                 leading to the separation of keratinocytes (Koch                ized by skin fragility with generalized superficial
                 et al. 1997). The loss of DSG1 function has so far              erosions and chronic inflammatory plaques and
                 not been analyzed in mice as it requires deletion               pruritus. Additional abnormalities include alo-
                 of all three DSG1 genes.                                        pecia and nail dystrophy (McGrath et al. 1997;
                      Desmosomal plaque proteins are equally                     Sprecher et al. 2004; McGrath 2005; McGrath
                 important for intercellular cohesion and me-                    and Mellerio 2010). Desmosomes in the skin
                 chanical stability: PG/JUP KO embryos died                      of patients were generally small with perturbed
                 as a result of severe heart defects with reduced                desmosome – keratin interactions. Severe der-
                 and structurally altered desmosomes (Bierkamp                   matitis, multiple allergies and metabolic wast-
                 et al. 1996; Ruiz and Birchmeier 1998). Al-                     ing (sinobronchial allergic mycosis, SAM) syn-
                 though b-catenin, the homologue of PG in                        drome (MIM615508) is a recently described
                 AJs, localized to desmosomes and could substi-                  genodermatosis caused by homozygous muta-
                 tute for PG in cadherin clustering, it failed to                tions in DSG1 or DSP. Dermatologic manifesta-
                 recruit normal levels of PKP1 and DSP to the                    tions comprise congenital erythroderma, striate
                 plaque (Bierkamp et al. 1999; Acehan et al.                     palmoplantar keratoderma (SPPK), skin ero-
                 2008). DSP KO embryos did not survive beyond                    sions, scaling, and hypotrichosis. Patients devel-
                 embryonic day 6.5 and displayed severe defects                  oped severe allergies, recurrent skin, and respi-
                 in tissue architecture, shaping of the embryo,                  ratory tract infections, indicating that loss of
                 and in anchoring keratin filaments to desmo-                    DSG1 interferes with skin barrier function (Sa-

                 4                Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                                                                                      Desmosomes and Keratins in Tissue Mechanics

                 muelov et al. 2013; Has et al. 2015). More re-                     and DSCs. Using analytical ultracentrifugation
                 cently, a point mutation in the keratin-binding                    and plasmon plasmon surface resonance reso-
                 domain of DSP was identified as causing SAM                        nance, these investigators determined KD-values
                 syndrome (McAleer et al. 2015). Nonsense, as                       for DSG and DSC homodimers, as well as het-
                 well as splice site mutations, have been observed                  erodimers. Whereas homodimers were very
                 in PG causing skin fragility, generalized epider-                  weak with KD values .400 mM, heterodimeric
                 molysis, palmoplantar keratoderma, and wool-                       pairs revealed KDs ranging from 3.6 to 43.9 mM,
                 ly hair (Pigors et al. 2011; Li et al. 2012). Ar-                  indicative of stronger adhesion than provided by
                 rhythmogenic cardiomyopathy (AC) is a rare                         E-cadherin (Harrison et al. 2016). These data
                 disease of the heart characterized by progres-                     correlate well with the role of AJs in mechano-
                 sive myocardial dystrophy with fibro-fatty re-                     sensing and one for desmosomes in conferring
                 placement. In the majority of cases, dominant                      stability. Interestingly, the strongest-binding
                 mutations have been identified in desmosomal                       pairs were Dsg1:Dsc1 and Dsg4:Dsc1, which
                 genes including DSP: ARVD8 (MIM607450);                            are expressed in the outermost layers of the epi-
                 PKP2: ARVD9 (MIM609040); DSG2: ARVD10                              dermis, whereas the basally expressed cadherins
                 (MIM610193); DSC2: ARVD11 (MIM610476);                             Dsg3:Dsc3 formed the weakest adhesive pair.
                 JUP: ARVD12 (MIM611528) (Al-Jassar et al.                          This correlates with the force-protective func-
                 2013; Cerrone and Delmar 2014; Calore et al.                       tion of the outer epidermal layers, although the
                 2015; Pilichou et al. 2016).                                       basal layers must allow for remodeling and re-
                     Desmosomes can also be affected in ac-                         generation, which is facilitated by weaker adhe-
                 quired diseases, which comprise epidermal au-                      sion. In agreement, preliminary experiments
                 toimmune disorders and infections (Stahley                         suggest an up-regulation of DSG1 on applica-
                 and Kowalczyk 2015). Pemphigus is a family                         tion of stretch to cultured keratinocytes (our
                 of diseases characterized by circulating autoan-                   own unpublished results).
                 tibodies that target desmosomal proteins and                            Similarly, PKPs -1 and -3 contribute differ-
                 compromise cell – cell adhesion (Amagai 2010;                      entially to intercellular cohesion. A comparison
                 Jennings et al. 2011; Amagai and Stanley 2012).                    of keratinocytes derived from the correspond-
                 Pemphigus autoantibodies are directed against                      ing KO mice confirmed that the loss of PKP1
                 the extracellular domains of desmosomal cad-                       interfered with formation of a stable epithelial
                 herins and are sufficient to cause the loss of                     sheet, whereas adhesion was not compromised
                 keratinocyte adhesion.                                             in PKP3 KO keratinocytes. Surprisingly, desmo-
                     Taken together, these studies clearly indicate                 somal PKP3 was not only much more dynamic
                 that desmosomes are essential for robust inter-                    compared with PKP1, but was able to destabi-
                 cellular cohesion and tissue integrity under me-                   lize PKP1-dependent desmosomes when over-
                 chanical strain.                                                   expressed (Keil et al. 2016). Collectively, these
                                                                                    data indicate that isoform expression has a con-
                 Why is Desmosomal Adhesion Stronger                                siderable influence on desmosome stability and
                 than AJ-Mediated Adhesion?                                         resistance to force and appears well suited to
                                                                                    adapt desmosomes to mechanical stresses.
                 Isoform-Dependent Composition
                 of Desmosomes Determines Adhesive
                 Strength                                                           Hyperadhesion is a Unique Feature
                                                                                    of Desmosomes
                 Although the differential expression of desmo-
                 somal cadherins and PKPs has been known for a                      In contrast to AJs, desmosomes can occur in
                 long time, experiments addressing their indi-                      two functionally distinct adhesive states, which
                 vidual and distinct contributions to desmoso-                      are distinguished on Ca2þ-depletion. In normal
                 mal adhesion have only recently emerged. Har-                      tissues, desmosomes adopt a Ca2þ-indepen-
                 rison et al. (2016) have systematically analyzed                   dent state, also referred to as hyperadhesion,
                 the structural basis of adhesive binding by DSGs                   which is crucial for stable intercellular cohesion

                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157      5
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                 and resistance to mechanical strain (Garrod                     a role of DSG2 in desmosome maturation and
                 2010; Thomason et al. 2010). In contrast, dur-                  in the switch to Ca2þ-independent adhesion
                 ing regeneration and wound healing, desmo-                      (Lowndes et al. 2014).
                 somes become Ca2þ-dependent resulting in                            Because an ordered intercellular zone re-
                 weaker intercellular cohesion, which allows for                 quires an ordered plaque, desmosomal plaque
                 tissue remodeling (Wallis et al. 2000; Kimura                   proteins are also important in regulating desmo-
                 et al. 2012). In cultured keratinocytes, desmo-                 somal hyperadhesion. PKP1 overexpression in-
                 somes depend on extracellular Ca2þ after their                  duced Ca2þ-independent hyperadhesive des-
                 formation but undergo a maturation process to                   mosomes. This prevented Pemphigus vulgaris
                 become hyperadhesive. Although the mecha-                       (PV)-IgG-mediated desmosome disruption
                 nism controlling formation of Ca2þ- insensitive                 (Tucker et al. 2014), indicative of a very stable
                 desmosomes is incompletely understood, one                      adhesive state. Moreover, PKP1 enhances des-
                 model predicts that a highly organized arrange-                 mosome size and number by laterally interact-
                 ment of the DSG and DSC extracellular do-                       ing with DSP in agreement with a postulated role
                 mains is induced (Thomason et al. 2010), which                  of plaque proteins in mediating cadherin spac-
                 correlates with a wider intercellular space and                 ing (Kowalczyk et al. 1999; Hatzfeld et al. 2000;
                 the presence of a midline observed in electron                  Bornslaeger et al. 2001; Hatzfeld 2007). Thus,
                 microscopic images, whereas Ca2þ-dependent                      the desmosomal cadherins, as well as the plaque
                 desmosomes appear to lack this midline (Gar-                    protein PKP1, cooperate to provide strong ad-
                 rod 2010).                                                      hesion and stability under mechanical strain.
                     Based on structural analyses of DSG2, Tariq
                 et al. (2015) proposed that the flexibility of des-             What are the Upstream Signals that Modulate
                 mosomal cadherin ectodomains allows them to                     Desmosomal Adhesion?
                 adopt a conformation that facilitates desmo-
                                                                                 Cross Talk Desmosomes-AJ
                 some plasticity. The desmosomal plaque would
                 contribute to plasticity by introducing spacings                Epithelial junction assembly, maturation, and
                 between desmosomal cadherins and preventing                     maintenance occur through sequential process-
                 ectodomain cis-interactions. The distance be-                   es that are differentially regulated (Keil et al.
                 tween cadherins and the lack of cis-interactions                2016). Earlier work suggested a hierarchical
                 facilitates conformational changes needed to                    process in which AJs form before desmosomes.
                 adopt the ordered architecture in the Ca2þ-in-                  However, increasing evidence indicates that
                 dependent state. Depletion of DSC2 prevented                    newly formed contacts contain components of
                 the acquisition of hyperadhesion in keratino-                   AJs and desmosomes, and sorting into distinct
                 cytes (Kurinna et al. 2014), although it is not                 junctional complexes occurs on junction mat-
                 known whether this is a unique feature of DSC2                  uration in epithelial cells (Green et al. 2010; Keil
                 or shared by all DSCs and DSGs. The fact that a                 et al. 2016).
                 DSC2 mouse KO did not reveal any phenotypic                         PG was the first protein identified in desmo-
                 abnormalities (Rimpler 2014) suggests that hy-                  somes and AJs based on colocalization studies
                 peradhesion does not critically depend on the                   and its coimmunoprecipitation with E-cadherin
                 DSC2 isoform. In an attempt to characterize the                 (Cowin et al. 1986; McCrea et al. 1991). Gosavi
                 combinatorial roles of DSCs and DSGs in des-                    et al. (2011) showed that PKP3, PG, and E-cad-
                 mosome assembly, micropatterned substrates                      herin are present at the cell border when cells are
                 were used to uncouple desmosome assembly                        grown in media with low concentrations of Ca2þ
                 from other cell contacts (Lowndes et al. 2014).                 and suggested a model in which PG and E-cad-
                 In this assay, DSC2, but not DSG2, formed                       herin recruit PKP3 to the cell border to initiate
                 Ca2þ-dependent homophilic bonds, indicating                     desmosome formation. A role of PKP3 in coor-
                 that DSC2 was required for desmosome assem-                     dinating AJ and desmosome formation was fur-
                 bly. In contrast, DSG2 formed Ca2þ-indepen-                     ther supported by the finding that PKP3 locali-
                 dent heterophilic bonds with DSC2, suggesting                   zation at the plasma membrane occurs together

                 6                Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                                                                                      Desmosomes and Keratins in Tissue Mechanics

                 with E-cadherin and precedes other desmo-                          Mechanosensitive Signaling Pathways in the
                 somal proteins, including PKP1 and DSP (Keil                       Control of Desmosomes
                 et al. 2016). At the molecular level, PKP3 forms a
                 complex with the Rap1 GTPase, promoting its                        So far, little is known about how desmosome
                 activation and facilitating desmosome assembly                     gene transcription and isotype expression are
                 (Todorovic et al. 2014). Moreover, PKP3 was                        controlled and how mechanical stimuli direct
                 required for AJ maturation and sealing. These                      these processes. Several recent studies have
                 findings reveal PKP3 as a coordinator of desmo-                    addressed the question how external mechani-
                 some assembly and AJ maturation through its                        cal forces are transmitted into chemical signals.
                 association with Rap1 (Todorovic et al. 2014).                     To induce changes in the cell’s gene expression
                                                                                    program, signaling molecules regulated by
                                                                                    external forces must be translocated from the
                 Actin and Desmosomes                                               cytoplasm into the nucleus and nucleocytoplas-
                 Formation of AJs but not desmosomes coin-                          mic shuttling has been suggested as a common
                 cides with an increase in the apparent stiffness                   theme in mechanotransduction (Sharili and
                 of cell monolayers reflecting the generation of a                  Connelly 2014). The SRF (serum-response fac-
                 tissue-level tension (Harris et al. 2014). Tension                 tor) and YAP (Yes-associated protein)/TAZ
                 rapidly increases with a maximum at 150 min,                       (transcriptional coactivator with PDZ-binding
                 which correlates with the initiation of desmo-                     motif ) pathways are known mediators of this
                 some assembly. Although desmosomes are as-                         process in multiple cell types, including kerati-
                 sociated with IFs and according to current views                   nocytes.
                 are unable to generate tension, their assembly                          SRF transcriptional activity is regulated by
                 requires actin microfilaments (Pasdar and Li                       actin and RhoA and its cofactors MRTF (myo-
                 1993). It is, therefore, possible that AJ-associat-                cardin-related transcription factor) -A and -B.
                 ed actomyosin-dependent force generation is an                     Monomeric actin sequesters MRTFs, which are
                 important signal for desmosome formation al-                       released on actin polymerization and accumu-
                 though direct experimental evidence remains to                     late in the nucleus where they interact with SRF
                 be adduced.                                                        to stimulate target gene expression. Intercellular
                      The AJ protein p120 plays an essential role                   tension can increase actin polymerization at AJs
                 in limiting actomyosin-dependent tension gen-                      and drive MRTF-A translocation into the nu-
                 erated at AJ as shown by uncoupling p120’s                         cleus (Gomez et al. 2010). SRF and MRTFs co-
                 cadherin-stabilizing and RhoA-suppressing ac-                      activate immediate early genes, such as JunB,
                 tivities. Removing p120’s Rho-suppressing ac-                      and adhesion-related genes, including vinculin.
                 tivity lead to excessive actomyosin contractility                  Mice with a keratinocyte-specific SRF-KO re-
                 along the vertical axis of cells and disrupted the                 vealed severe intercellular gaps between kerati-
                 integrity of the apical surface, irrespective of E-                nocytes as the most striking pathophysiology
                 cadherin stability (Yu et al. 2016). Interestingly,                (Koegel et al. 2009). The number of desmo-
                 p120 has been described to associate with des-                     somes was significantly lower in mutant com-
                 mosomes during their formation in a Ca2þ-                          pared with control mouse skin, whereas the size
                 shift experiment (Kanno et al. 2008).                              of individual desmosomes was unaltered. How-
                      Desmosomes can also influence actin orga-                     ever, a direct regulation of desmosomal genes
                 nization: Loss of any of the PKPs from human                       was not supported by transcription profiles of
                 or mouse keratinocytes results in changes in                       SRF KO skin samples compared with control
                 cortical actin organization (Godsel et al. 2010;                   skin (Koegel et al. 2009). In contrast, loss of
                 Keil et al. 2016). However, it is not clear whether                MRTF-A reduced the levels of DSG1 messenger
                 PKPs regulate RhoA activity and stress fiber for-                  RNA (mRNA) in keratinocytes, suggesting that
                 mation directly or indirectly, by influencing the                  DSG1 is directly regulated by the SRF/MRTF-A
                 localization or activity of a Rho GEF or a Rho                     complex (Dubash et al. 2013). Another experi-
                 GAP (Godsel et al. 2010).                                          ment showed that MRTF-A and SRF were

                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157      7
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                 recruited to cis-regulatory elements of the PKP2                also respond to the rigidity of the extracellular
                 gene to regulate its expression (Leitner et al.                 matrix, cell geometry, cell density, cell polarity,
                 2011). Taken together, these data suggest that                  and the status of the actin cytoskeleton. Sensing
                 RhoA/MRTF-A signaling in keratinocytes af-                      of substrate stiffness by YAP/TAZ depends on
                 fects the expression of desmosomal proteins di-                 the tension of the actin cytoskeleton (Zhang
                 rectly and indirectly although the regulation via               et al. 2011; Elbediwy et al. 2016). The AJ protein
                 SRF/MRTF-A in response to mechanical strain                     a-catenin limits YAP activity by modulating its
                 has not been directly shown.                                    interaction with 14-3-3 (Kanai et al. 2000; Schle-
                     The transcription factor AP-1 mediates gene                 gelmilch et al. 2011; Sambandam et al. 2015).
                 expression in response to a variety of extracel-                YAP/TAZ mediate their function in controlling
                 lular stimuli and is activated by SRF/MRTF sig-                 gene expression through interaction with TEAD
                 naling (Wang et al. 2015). Microarray analysis                  transcription factors, which drive the expression
                 performed on mechanical stretched and normal                    of proliferative genes (Zhang et al. 2011). Al-
                 human skin revealed an up-regulation of AP-1                    though YAP activation accelerates proliferation
                 in mechanically stretched skin (Yang et al.                     and suppresses differentiation of mouse kerati-
                 2011). Other stimuli of AP-1 include growth                     nocytes, which correlates with altered desmo-
                 factor signaling via mitogen-activated protein                  some number, size, and composition, a role of
                 (MAP) kinases, which are also activated in re-                  YAP/TAZ/TEAD in the transcriptional regula-
                 sponse to mechanical stress (Kippenberger et al.                tion of desmosomal genes has not been directly
                 2000). AP-1 acts as a homodimeric or hetero-                    addressed. However, a large scale ChIP-seq anal-
                 dimeric transcription factor composed of mem-                   ysis of Tead4 target genes in ECC1 endometrial
                 bers of the Fos (c-Fos, FosB, Fra-1, and Fra-2)                 cells identified DSCs 1-3, DSG1, DSP, PG, and
                 and Jun (c-Jun, JunB, and JunD) families. Jun is                PKPs -1 and -2 as putative Tead4 target genes
                 regarded as a positive regulator of keratinocyte                (Liu et al. 2016).
                 proliferation/differentiation during develop-
                 ment and in skin cancer through its direct tran-
                 scriptional effect on epidermal growth factor                   ROLE OF IFs IN MECHANICAL STABILITY
                 receptor (EGFR) expression. In contrast, JunB
                                                                                 Composition, Assembly, and Organization
                 can antagonize proliferation of keratinocytes.
                                                                                 of Keratin Filaments
                 AP-1 target genes include keratins (Yates and
                 Rayner 2002).                                                   IFs can be assembled from a superfamily of
                     The Hippo signaling pathway is another reg-                 approximately 70 proteins that form cell-type-
                 ulator of mechanotransduction. It regulates or-                 specific cyto- and nucleoskeletal arrays in the
                 gan size, tissue regeneration, and stem cell re-                majority of multicellular animal species. Based
                 newal (Zhang et al. 2011; Barry and Camargo                     on sequence homology and expression, IFs are
                 2013). Recently, mechanical signals were found                  grouped into six classes, all of which except lam-
                 to be transduced by the two mediators of the                    ins form predominantly cytoplasmic assemblies
                 Hippo pathway, the transcriptional coactivators                 (Schweizer et al. 2006; Hobbs et al. 2016). Here,
                 YAP and TAZ (Dupont et al. 2011). In the skin,                  we focus on epithelial keratins to examine their
                 YAP is required for epidermal barrier formation,                contribution to the mechanical resilience of ep-
                 hair follicle development, and maintenance of                   ithelia in conjunction with desmosomes, al-
                 the epidermal stem cell compartment (Zhang                      though desmin and vimentin also interact with
                 et al. 2011; Barry and Camargo 2013; O’Neill                    desmosomes in cardiac tissue, meningiomal,
                 2015). Overexpression of YAP in transgenic                      and arachnoidal cells in which they fulfill anal-
                 mice promotes squamous cell carcinoma for-                      ogous functions (Kartenbeck et al. 1983, 1984).
                 mation (Chan et al. 2011; Jia et al. 2016), where-                   Keratin assembly begins with heterodimeri-
                 as its knockdown inhibits cutaneous wound                       zation of type I and II keratin molecules, fol-
                 healing (Lee et al. 2014), suggesting a central                 lowed by formation of antiparallel tetramers.
                 role in regulating proliferation. YAP and TAZ                   The lateral association of four tetramers gener-

                 8                Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                                                                                      Desmosomes and Keratins in Tissue Mechanics

                 ates so-called unit-length filaments (ULFs),                       follicle, and are characterized by the expression
                 which longitudinally coalesce into mature fila-                    of KRT15 (Lyle et al. 1998; Watt 1998; Goldstein
                 ments with a propensity to bundle and organize                     and Horsley 2012). The vibrissae bulge harbors
                 into three-dimensional (3D) networks (Herr-                        two types of slow cycling stem cells, character-
                 mann et al. 2009; Koster et al. 2015; Herrmann                     ized by the expression of KRT5/15/17/19 and
                 and Aebi, 2016). Keratins show extensive se-                       KRT5/17, respectively, which display loose ker-
                 quence diversity, a feature that discriminates                     atin bundles in the former and tight bundles
                 them from actins and tubulins (Schweizer et                        in the latter configurations (Larouche et al.
                 al. 2006; Fletcher and Mullins 2010). This se-                     2008). Notably, in both subpopulations lacking
                 quence diversity is responsible for their relative                 KRT14, keratin network organization is differ-
                 affinities to each other and to associated pro-                    ent. The complexity of keratin expression sug-
                 teins, and enables isotype-specific posttrans-                     gests that keratins contribute to an intracellular
                 lational modifications (Hatzfeld and Franke                        epithelial niche, analogous to a stem cell niche
                 1985; Hofmann and Franke 1997; Snider and                          (Tumbar et al. 2004), that endows distinct epi-
                 Omary 2014). In humans and the mouse, 28                           thelial cells with unique micromechanical prop-
                 type I keratins (KtyI) and 26 type II keratins                     erties through formation of specifically tailored
                 (KtyII) genes are predominantly expressed as                       keratin-desmosome networks (Fig. 2).
                 pair-specific combinations of types I and II ker-
                 atin proteins, respectively, in epithelial cells and
                                                                                    Human Disease and Mouse Models
                 tissues. The major impact of keratins on cell
                 integrity and adhesion is most obvious in the                      The importance of keratins for providing me-
                 epidermis, in addition to embryonic epithelia                      chanical stress resilience in the epidermis is best
                 (Simpson et al. 2011; Bouameur and Magin                           documented by blistering and hyperkeratotic
                 2017). The basal compartment expresses the                         skin disorders exemplified by Epidermolysis
                 keratin pair KRT5/KRT14, organized in loose                        bullosa simplex (EBS; OMIM #131900), caused
                 bundles that extend from hemidesmosomes                            by mutations in KRT5 and KRT14 (Szeverenyi
                 and desmosomes. On terminal differentiation,                       et al. 2008). EBS is characterized by cytoplasmic
                 these keratinocytes disconnect from the ECM                        keratin aggregates, cytolysis of basal keratino-
                 accompanied by replacement of KRT5/KRT14                           cytes, and bullous lesions following mild trau-
                 with KRT1/KRT10. Depending on regional dif-                        ma to the skin. Although it is recognized that
                 ferences in the epidermis, additional keratins                     the pathomechanisms contributing to EBS and
                 become expressed. At sites of high mechanical                      additional keratinopathies are more complex
                 strain, such as palms and soles, the keratins                      than originally considered (Coulombe and Lee
                 KRT1/KRT10 are supplemented by keratins                            2012; Roth et al. 2012; Bohnekamp et al. 2015;
                 KRT2e and KRT9 (Moll et al. 2008). Analogous                       Hobbs et al. 2016; Kumar et al. 2016), it is evi-
                 to desmosomes, keratin concentration increases                     dent that loss of an intact keratin cytoskeleton
                 from 40 mg/ml in basal keratinocytes to prob-                      renders keratinocytes fragile on mild physical
                 ably twice as much in terminally differentiated                    stress, shown by KRT5 and KRT14 KO mice
                 keratinocytes (Sun and Green 1978; Feng et al.                     (Lloyd et al. 1995; Peters et al. 2001). Of note,
                 2013). The above keratin expression pattern var-                   even mutations causing severe disease do not
                 ies, for example, during epidermal injury, which                   prevent formation of long keratin intermediate
                 triggers the rapid induction of KRT6, -16, and                     filaments (KIFs) in vitro (Herrmann et al.
                 -17 at the wound edge at the expense of KRT1/                      2002), suggesting that mutations and physical
                 10, accompanied by fewer and less adhesive des-                    stress act at the level of keratin bundling, net-
                 mosomes (Garrod and Chidgey 2008; Simpson                          work organization, dynamics, or by affecting
                 et al. 2011). KRT6, KRT16, and KRT17 are also                      association with other proteins. Indeed, the
                 expressed in hair follicles and the nails (Moll et                 most frequent KRT14 Arg125 mutation com-
                 al. 2008). Epidermal stem cells are located in                     promises desmosome adhesion (Russell et al.
                 protected niches, such as the bulge of the hair                    2004; Homberg et al. 2015).

                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157       9
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                      Deletion and/or mutation of other keratin                  (Gallicano et al. 1998; Vasioukhin et al. 2001;
                 isotypes expressed in different epidermal com-                  Sumigray and Lechler 2012). Moreover, DSP
                 partments, either in the mouse or in humans,                    and DSG1 mutations can give rise to SPPK
                 did not cause cytolysis and skin blistering to the              (OMIM #612908, #148700, respectively), a hy-
                 same extent as KRT5 and KRT14, but was typ-                     perkeratotic skin condition with fewer or small-
                 ified by altered cell and tissue growth, barrier,               er desmosomes in the suprabasal epidermis and
                 and immune defects (Reichelt et al. 2001;                       perinuclear accumulation of KIF in DSP-asso-
                 McGowan et al. 2002; Lessard and Coulombe                       ciated SPPK (Wan et al. 2004). Conversely, de-
                 2012; Roth et al. 2012; Fischer et al. 2014, 2016;              letion of keratins reduced DSP deposition at the
                 Fu et al. 2014; Kumar et al. 2016; Bouameur and                 plasma membrane (Loranger et al. 2006; Vi-
                 Magin 2017). Transgenic expression of a bun-                    jayaraj et al. 2009). Distinct steps during desmo-
                 dling-competent KRT5/KRT8 chimaera par-                         some assembly also require a transient but stable
                 tially rescued KRT52/2 mice, whereas expres-                    interaction between DSP and KIF in the cyto-
                 sion of bundling-deficient KRT8 did not,                        plasm (Godsel et al. 2005; Hobbs and Green
                 providing strong evidence that, in addition to                  2012; Albrecht et al. 2015).
                 keratin abundancy, isotype-specific properties                      Keratins interact with the plectin and DSP
                 and keratin network organization are crucial                    carboxy termini through segments of their rod
                 contributors to keratin-mediated stress resil-                  domain (Meng et al. 1997; Fontao et al. 2003;
                 ience (Alvarado and Coulombe 2014).                             Bouameur et al. 2014). Association with kera-
                      The known redundancy of keratin ex-                        tins is regulated by serine phosphorylation and
                 pression in part explains lack of similarly severe              arginine methylation of the DSP tail, involving a
                 defects in other epithelial compartments and                    coordinated activity of glycogen synthase kinase
                 complicates the analysis of isotype-specific                    3 (GSK3) and protein arginine methyltransfer-
                 functions (Reichelt et al. 2001; McGowan et al.                 ase 1 (PRMT1). Inhibition of GSK3 or PRMT1
                 2002; Lessard and Coulombe 2012; Roth et al.                    delayed desmosome assembly and enhanced
                 2012). To overcome the latter, mice lacking the                 DSP–KIF interactions in the cytoplasm (Al-
                 entire KtyI and KtyII genes in their epidermis                  brecht et al. 2015). In the absence of keratins,
                 were generated, in addition to mice lacking both                using keratinocytes from keratin-deficient mice
                 KRT1 and KRT10. Such mice developed a fully                     (Vijayaraj et al. 2009), DSP phosphorylation
                 stratified epidermis but died perinatally because               was elevated and desmosomes were endocy-
                 of extensive epidermal damage (Wallace et al.                   tosed at accelerated rates. Such desmosomes
                 2012; Bar et al. 2014; Kumar et al. 2015). Most                 were unable to render epithelial sheets stable
                 importantly, these mice showed diminished in-                   on rotational stress (Kroger et al. 2013). Reex-
                 tercellular adhesion and significantly smaller                  pression of KRT5 and KRT14 reconstituted
                 desmosomes. This was accompanied by accu-                       properly formed desmosomes and shear-resis-
                 mulation of desmosomal proteins in the cyto-                    tant intercellular adhesion. Preliminary evi-
                 plasm, highlighting that in vivo interaction with               dence indicates that sequestration of protein ki-
                 keratins was required for the maintenance of                    nase C-a (PKCa) by the scaffold protein Rack1
                 functionally intact desmosomes.                                 and KRT5/14 contribute to the stabilization of
                                                                                 desmosomes (Kroger et al. 2013), whereas PKP3
                                                                                 may recruit PKCa to abrogate hyperadhesion
                 Interdependence of Keratins and
                                                                                 (Keil et al. 2016). In agreement, loss of PKCa
                 Desmosomes
                                                                                 in mice delays reepithelialization following
                 Desmosomes have been recognized early on as                     wounding and is accompanied by maintenance
                 important sites for KIF formation and organi-                   of hyperadhesive desmosomes (Thomason et al.
                 zation in vivo (Jackson et al. 1980; Bologna et al.             2012).
                 1986; Schwarz et al. 2015), and genetic deletion                    Reepithelialization requires not only altered
                 of DSP can cause extensive reorganization or                    adhesion of keratinocytes at the wound edge
                 collapse of KIF, depending on the cell type                     (Shaw and Martin 2009) but also diminished

                 10               Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                                                                                      Desmosomes and Keratins in Tissue Mechanics

                 contacts of keratins to desmosomal proteins,                       well accepted, the contribution of keratins to the
                 along with elevated expression of KRT6/16/17                       resilience of epithelia against various types of
                 and a decrease in KRT1/10 (Patel et al. 2006). In                  deformation remained unknown. Unlike other
                 agreement, cells expressing KRT6 or KRT17                          cytoskeletal elements, IF networks are less rigid
                 show elevated, PKCa-mediated desmosome                             at low shear strain but harden at high strains and
                 disassembly and subsequent destabilization of                      resist breakage, indicating that they are crucial
                 epithelial sheets. In contrast, KRT5 or KR14                       for the maintenance of tissue integrity (Janmey
                 supported stable desmosomes, suggesting that                       et al. 1991, 2013). Using atomic force microsco-
                 expression of “wound healing” keratins weakens                     py (AFM)-based single-cell compression to ex-
                 intercellular adhesion. Further studies suggest                    amine resistance against external pressure and
                 that the type II keratin KRT5 is a major deter-                    global rupturing forces, keratinocytes were 6 –
                 minant of desmosome stability (Loschke et al.                      70 times stiffer than most other cell types. Selec-
                 2016). In addition to PKCa, p38 MAP kinase                         tive disruption of actin filaments and microtu-
                 (MAPK) signaling downstream from the serine                        bules did not significantly alter keratinocyte
                 protease inhibitor SPINT1 participates in the                      mechanics (Lulevich et al. 2010). However, ap-
                 interaction of keratins and desmosomal pro-                        plication of AFM to keratin-deficient individual
                 teins (Kawaguchi et al. 2015).                                     keratinocytes revealed a significant softening
                      Thus, available data provide strong evidence                  of keratin-deficient cells (Ramms et al. 2013).
                 for an interdependence of keratins and DSP                         Magnetic tweezer experiments additionally
                 regulated by posttranslational modifications.                      showed a major contribution of keratins to vis-
                 The casein kinase I (CK-1a) scaffold protein                       coelastic cell properties. Similar conclusions
                 FAM83H regulates keratin networks by recruit-                      were reached using a microfluidic optical
                 ing CK-1a directly to keratins (Kuga et al.                        stretcher (Seltmann et al. 2013). To relate these
                 2013). It may be additionally involved in mem-                     findings to functional consequences, invasion
                 brane localization of desmosomal proteins and                      and 3D growth assays were performed with nor-
                 in regulating desmosome – keratin interactions                     mal and keratin-deficient keratinocytes, reveal-
                 (Kuga et al. 2016). Whether FAM83H activity                        ing higher invasiveness in the latter. However,
                 toward keratins or desmosomal proteins is                          this came at the price of increased disruption
                 coordinated with force-dependent conforma-                         of cells during invasion, suggesting that certain
                 tional changes in desmosomal proteins remains                      levels of keratins or other IF proteins are re-
                 currently unknown. In Xenopus mesendoderm                          quired to sustain the invasive process (Cheung
                 cells, increased tension at P-cadherin-contain-                    et al. 2013; Seltmann et al. 2013).
                 ing junctions recruits PG/JUP to these sites. In                        Beyond single cells, very little is known
                 turn, PG/JUP recruits KIF to these junctions to                    about the contribution of keratin architecture
                 reorganize and reinforce the keratin cytoskele-                    to tissue mechanics. A recent AFM-based study,
                 ton. Through this sequence of events, local forc-                  using the human hair follicle as a model system,
                 es from neighboring cells mediate keratin reor-                    showed stiffening of the soft keratinocyte matrix
                 ganization required for coordinated cell                           at the base of the hair follicle 360-fold, from
                 behavior (Weber et al. 2012).                                      30 kPa to 11 MPa along the first millimeter of
                                                                                    the follicle. This stiffening coincided with an
                                                                                    increased thickness of keratin macrofibrils and
                 Keratins as Main Determinants for the
                                                                                    their orientation. The continued stiffening like-
                 Mechanical Integrity of Keratinocytes and
                                                                                    ly is because of increasing network orientation,
                 Tissues
                                                                                    compaction, and mechanical reinforcement of
                 The mechanical properties of epithelial cells                      the keratin macrofibrils by disulfide cross-links.
                 largely depend on actin filaments, microtubules,                   This study links changes in the architecture and
                 and KIF (Fletcher and Mullins 2010; Koster et al.                  molecular structure of keratin macrofibrils to
                 2015). Although the contribution of actin fila-                    the local mechanical behavior at a tissue scale
                 ments and microtubules to these properties is                      level (Bornschlogl et al. 2016).

                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157      11
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                 Impact of Mechanical Stretch on the                             response of keratins, affecting predominantly
                 Desmosome –Keratin Complex                                      the peripheral keratin network (Fois et al.
                                                                                 2013). A set of studies has investigated bio-
                 Exposure of keratinocytes to stretch results                    physical and physiological consequences of
                 in rapid and transient induction of MAPK                        hyperphosphorylation on normal and mutant
                 ERK1/2 and SAP kinases (Kippenberger et al.                     keratins. The bioactive lipid sphingosylphos-
                 2000; Yano et al. 2004), accompanied by altered                 phorylcholine (SPC) activates JNK and Erk ki-
                 keratin expression, increased keratin phosphor-                 nases, mediating keratin reorganization through
                 ylation and reorganization of keratin networks                  phosphorylation of KRT8 at Ser431 and KRT18
                 (Yano et al. 2004; Snider and Omary 2014). At                   at Ser52 in pancreatic tumor cells. The resulting
                 least in keratinocytes, mechanical stretching                   perinuclear keratin reorganization affects the
                 acts via induction of calcium influx, EGFR                      viscoelasticity of metastatic cancer cells and pro-
                 phosphorylation, and ERK1/2 activation                          motes tumor invasion (Beil et al. 2003). Me-
                 (Yano et al. 2004). In this setting, the established            chanically stretching of keratinocytes expressing
                 role of DSG1 in EGFR signaling has not yet been                 EBS-like keratin mutations triggered a progres-
                 examined, thus, it remains unknown whether                      sive disassembly of desmosomes and weakened
                 keratin reorganization is mediated by desmo-                    intercellular adhesion (Russell et al. 2004; Hom-
                 somes or affects them (Harmon et al. 2013).                     berg et al. 2015). Thus, keratin reorganization
                 On mechanical stretch, keratinocytes expressing                 on posttranslational modifications might con-
                 the disease mutation KRT10 Arg156His show                       tribute to the invasive properties of metastatic
                 stronger activation of p38 MAPK compared                        tumor cells and to altered adhesion in skin dis-
                 with control transfectants, which ultimately                    orders (Beil et al. 2003; Russell et al. 2004; Hom-
                 might drive hyperproliferation typical of the                   berg et al. 2015; Loschke et al. 2016).
                 corresponding keratinopathy (Obarzanek-Fojt
                 et al. 2011). In lung epithelial cells, shear stress,
                 but not stretch, caused hyperphosphorylation                    Intrinsic Keratin Properties and Associated
                 and disassembly of KRT8 and KRT18-contain-                      Proteins as Determinants of Network
                 ing IF, regulated by PKC-d (Ridge et al. 2005).                 Organization and Stress Resilience
                 Blocking the shear stress – mediated keratin hy-                The organization of cytoskeletal networks de-
                 perphosphorylation and reorganization de-                       pends on their intrinsic properties and their
                 creased cell viability and increased apoptosis.                 associated proteins, and determines the com-
                 Notably, shear stress induced bundling of KIF                   prehensive array of functions that they perform
                 into thick fibrils. In this study, shear stress – me-           (Fletcher and Mullins 2010). Bundling of indi-
                 diated phosphorylation of KRT18 at Ser33 was a                  vidual filaments into fibers, an intrinsic keratin
                 prerequisite for keratin network reorganization                 property (Lee and Coulombe 2009), is seen in
                 to alter cells’ mechanical properties and sustain               most epithelia, evident in the epidermis and in
                 their integrity (Flitney et al. 2009; Sivaramak-                hair follicles, whereas simple epithelia show less
                 rishnan et al. 2009).                                           dense arrays. Here, an interesting contributor to
                      To understand how phosphorylation affects                  IF organization lies in their capacity to form
                 keratin reorganization, select phosphomutants                   disulfide bonds. With the exception of KRT8,
                 of KRT8 and KRT18 were characterized for their                  KRT18, and KRT19, all other human and
                 network mechanical properties using rheology                    mouse keratins contain variable numbers of cys-
                 and electron microscopy. Phosphokeratin-con-                    teine residues. Following the observation that a
                 taining networks showed reduced intraconnec-                    subset of keratins becomes disulfide-cross-
                 tivity and resulted in mechanically weaker and                  linked on terminal differentiation in vivo, ho-
                 more deformable networks in vitro. This ap-                     motypic disulfide bonds involving three Cys res-
                 peared to result from the formation of shorter                  idues in KRT14 and one in K10 were identified
                 mutant filaments (Deek et al. 2016). In cultured                in cultured keratinocytes and mouse epidermis,
                 cells, hyperphosphorylation alters the stretch                  in addition to interkeratin disulfide-bonded

                 12               Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                                                                                      Desmosomes and Keratins in Tissue Mechanics

                 KRT5/14 species (Lee et al. 2012; Feng and Cou-                    domains may uncover the hidden SH3 domain
                 lombe 2015; Bunick and Milstone, 2016). Avail-                     (Osmani and Labouesse 2015).
                 able data point to multiple roles of disulfide-                         Very little is known about the interaction of
                 bonding on keratin assembly, dynamics, and                         keratins with actin and actomyosin, which is
                 network organization. Intriguingly, disulfide                      crucial during morphogenesis, cell polarity,
                 bonding of K14 coincided with formation of                         cell migration, and invasion. In a study aimed
                 networks enriched in the nuclear periphery, a                      to normalize mutation-disrupted keratin net-
                 region reactive to mechanical cues (Feng and                       works in hepatocytes, the multikinase inhibitor
                 Coulombe 2015; Wallrath et al. 2016). Disulfide                    PKC412 was found to perform this function by
                 bonding may also alter bending and buckling of                     enhancing nonmuscle myosin heavy chain-IIA
                 keratin networks, which occur on compressive                       (NMHC-IIA) interaction with KRT8 and KRT
                 intracellular forces (Nolting et al. 2014). Finally,               18 through inhibiting NMHC-IIA phosphory-
                 disulfide bonds in keratins not only alter keratin                 lation (Kwan et al. 2015). Solo (ARHGEF40) is a
                 organization and dynamics on mechanical sig-                       RhoA-targeting guanine nucleotide exchange
                 nals but could tie them to the highly dynamic                      factor (GEF) involved in cyclical stretch-induced
                 cellular redox network controlled by the antiox-                   human endothelial cell reorientation and con-
                 idant transcription factor Nrf2 (Schafer et al.                    vergent extension cell movement in zebrafish
                 2012).                                                             gastrula and binds to KRT8/18 IF. Knockdown
                      In addition to posttranslational modifica-                    of Solo suppresses tensile force– induced stress
                 tions, the small heat shock protein Hsp27 and                      fiber reinforcement and RhoA activation. Thus,
                 proteins of the plakin and S100 families regulate                  the interplay between Solo and KRT8/18 plays a
                 keratin assembly, bundling, and network orga-                      crucial role in tensile force– induced RhoA acti-
                 nization (Windoffer et al. 2011; Kayser et al.                     vation and consequent actin cytoskeletal rein-
                 2013; Bouameur et al. 2014; Lesniak and Grac-                      forcement (Fujiwara et al. 2016).
                 zyk-Jarzynka 2015). Plectin is a plakin protein
                 that links keratin bundling to MAPK signaling.
                 In contrast to prediction, genetic deletion of                     CONCLUDING REMARKS
                 plectin rendered cells more susceptible to me-
                                                                                    Desmosomes as Mechanosensors?
                 chanical stress, despite an increase in keratin
                 bundling (Osmanagic-Myers et al. 2006). The                        As mentioned above, AJs are considered as the
                 latter is consistent with unaltered mechani-                       mechanosensing unit in intercellular force
                 cal properties of plectin knockdown cells as                       transduction and distribution in multicellular
                 assessed by indentation analyses using AFM                         tissues. It remains an open question whether or
                 and by displacement analyses of cytoplasmic                        not desmosomes can function as mechanosen-
                 superparamagnetic beads using magnetic twee-                       sors. Although desmosomes are composed of
                 zers (Moch et al. 2016). To investigate the trans-                 structurally related sets of proteins, including
                 mission of forces to the nucleus, which is crucial                 cadherin and armadillo family members, a di-
                 for nuclear function, a recent study examined                      rect homologue of the key mechanosensor in
                 plectin’s role in controlling nuclear morphology                   AJs, a-catenin is lacking. Instead, desmosomes
                 via keratins. On plectin knockdown, actomyo-                       contain the cytoskeletal linker protein DSP with
                 sin-dependent nuclear deformation occurred,                        an important role in lateral clustering of cadher-
                 whereas direct interactions between keratins                       ins and in linking KIFs to the junction. Its ami-
                 and the nuclear envelope were not required. In-                    no-terminal domain interacts with PG/JUP
                 stead, plectin down-regulation reduced KIF                         and PKPs, and is essential for DSP clustering
                 density in the nuclear perimeter (Almeida                          and determining desmosome size. This domain
                 et al. 2015). Possibly, plectin acts as a mechano-                 contains spectrin repeats homologous to the
                 sensor as it contains a cryptic SH3 domain in the                  spectrin repeats found in the related plakin fam-
                 fifth spectrin repeat of its plakin domain. Fol-                   ily protein, plectin (Choi and Weis 2011). Spec-
                 lowing mechanical stress, unfolding of spectrin                    trin repeat domains undergo only moderate

                 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157     13
Downloaded from http://cshperspectives.cshlp.org/ on January 22, 2022 - Published by Cold Spring Harbor Laboratory Press

                 M. Hatzfeld et al.

                 bending under mechanical stress, but repeats                    conserved in epithelial cells and between PKP
                 were shown to unfold individually at low pull-                  isoforms because the majority of published YAP
                 ing forces (Rief et al. 1999). Thus, it is possible             targets from mouse keratinocytes (Liu et al.
                 that the DSP-amino-terminal spectrin repeat                     2016) appeared down-regulated in PKP1 KO
                 domain undergoes force-induced unfolding of                     keratinocytes (our own unpublished results).
                 individual spectrin repeats leading to confor-                  Taken together, these data indicate that desmo-
                 mational changes that might alter DSP– protein                  somes might regulate YAP/TAZ signaling. Be-
                 interactions. In such a scenario, DSP would act                 cause YAP and PG/JUP were coimmunopreci-
                 directly as a mechanosensor. Interestingly, sev-                pitated from human heart protein extracts,
                 eral disease-causing mutations cluster in this                  desmosomes could function along with a-cat-
                 region and are potentially linked to structural                 enin to recruit YAP to cell–cell contacts and
                 destabilization (Ortega et al. 2016). A central,                limit its transcriptional activity.
                 a-helical domain of DSP forms coiled coils
                 leading to homodimerization. In contrast to
                 the spectrin repeat domain, coiled-coils bend                   ACKNOWLEDGMENTS
                 easily but are rather resistant to stretching (Ada-
                 movic et al. 2008). The carboxy-terminal do-                    Deutsche Forschungsgemeinschaft (DFG) fund-
                 main of DSP interacts with keratins and consists                ing to the Hatzfeld and Magin laboratories is
                 of three plakin repeat domains. Whereas the                     gratefully acknowledged (Ha1791/8-1, Ha1791/
                 first plakin repeat domains are linked by just                  10-1, MA1316-15, MA1316-17, MA1316-19,
                 four amino acids, plakin repeat domains 2 and                   MA1316-21, INST 268/230-1).
                 3 are separated by a 154 amino acid linker (Kang
                 et al. 2016). Decoupling of the plakin domains
                 suggests that the hinge region might be extend-                 REFERENCES
                 ed and strained in an initial step, before the                  
                                                                                     Reference is also in this collection.
                 amino-terminal spectrin repeats undergo
                 force-induced conformational changes (Ortega                    Acehan D, Petzold C, Gumper I, Sabatini DD, Muller EJ,
                                                                                    Cowin P, Stokes DL. 2008. Plakoglobin is required for
                 et al. 2016). Therefore, the plakin domain may                     effective intermediate filament anchorage to desmo-
                 work as a molecular shock absorber that dissi-                     somes. J Invest Dermatol 128: 2665–2675.
                 pates elastic energy when cells are subjected to                Adamovic I, Mijailovich SM, Karplus M. 2008. The elastic
                                                                                    properties of the structurally characterized myosin II S2
                 external forces (Ortega et al. 2016).                              subdomain: A molecular dynamics and normal mode
                                                                                    analysis. Biophys J 94: 3779– 3789.
                                                                                 Albrecht LV, Zhang L, Shabanowitz J, Purevjav E, Towbin JA,
                 Desmosomes in Mechanotransduction                                  Hunt DF, Green KJ. 2015. GSK3- and PRMT-1-depen-
                                                                                    dent modifications of desmoplakin control desmopla-
                 As mentioned before, the Hippo mediators                           kin-cytoskeleton dynamics. J Cell Biol 208: 597–612.
                 YAP/TAZ may be involved in regulating desmo-                    Al-Jassar C, Bikker H, Overduin M, Chidgey M. 2013.
                 somes via the transcription factor Tead4 (Liu et                   Mechanistic basis of desmosome-targeted diseases. J
                                                                                    Mol Biol 425: 4006– 4022.
                 al. 2016). Recently, desmosome mutations in
                                                                                 Almeida FV, Walko G, McMillan JR, McGrath JA, Wiche G,
                 AC were linked to modulation of Hippo/YAP                          Barber AH, Connelly JT. 2015. The cytolinker plectin
                 signaling, suggesting that there might be a feed-                  regulates nuclear mechanotransduction in keratinocytes.
                 back/feedforward mechanism (Chen et al.                            J Cell Sci 128: 4475– 4486.
                 2014). In AC, levels of phospho-YAP were in-                    Alvarado DM, Coulombe PA. 2014. Directed expression of a
                                                                                    chimeric type II keratin partially rescues keratin 5-null
                 creased compared with normal human hearts,                         mice. J Biol Chem 289: 19435– 19447.
                 and phospho-YAP was recruited to the junc-                      Amagai M. 2010. Autoimmune and infectious skin diseases
                 tions. In agreement, RNA sequencing, quanti-                       that target desmogleins. Proc Jpn Acad Ser B Phys Biol Sci
                                                                                    86: 524– 537.
                 tative polymerase chain reaction, and reporter
                                                                                 Amagai M, Stanley JR. 2012. Desmoglein as a target in skin
                 assays all showed suppressed TEAD activity in                      disease and beyond. J Invest Dermatol 132: 776– 784.
                 HL-1 myocytes with a PKP2 knockdown (Chen                       Bar J, Kumar V, Roth W, Schwarz N, Richter M, Leube RE,
                 et al. 2014). We suppose that this mechanism is                    Magin TM. 2014. Skin fragility and impaired desmo-

                 14               Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029157
You can also read