<<

Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Adherens Junctions and Coordinate Mechanics and Signaling to Orchestrate Tissue Morphogenesis and Function: An Evolutionary Perspective

Matthias Rübsam,1,4 Joshua A. Broussard,2,4 Sara A. Wickström,3 Oxana Nekrasova,2 Kathleen J. Green,2 and Carien M. Niessen1

1University of Cologne, Department of Dermatology, Cologne Excellence Cluster on Stress Responses in Aging Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC) at the CECAD Research Center, 50931 Cologne, Germany 2Northwestern University Feinberg School of Medicine, Departments of Pathology and Dermatology, the Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois 60611 3Paul Gerson Unna Group, Skin Homeostasis and Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany Correspondence: [email protected]; [email protected]

Cadherin-based adherens junctions (AJs) and desmosomes are crucial to couple intercellular adhesion to the or intermediate filament , respectively. As such, these intercellular junctions are essential to provide not only integrity to epithelia and other tissues but also the mechanical machinery necessary to execute complex morphogenetic and ho- meostatic intercellular rearrangements. Moreover, these spatially defined junctions serve as signaling hubs that integrate mechanical and chemical pathways to coordinate tissue archi- tecture with behavior. This review takes an evolutionary perspective on how the emergence of these two essential intercellular junctions at key points during the evolution of multicellular animals afforded metazoans with new opportunities to integrate adhesion, cytoskeletal dy- namics, and signaling. We discuss known literature on cross-talk between the two junctions and, using the skin as an example, provide a model for how these two junctions function in concert to orchestrate tissue organization and function.

pithelial barrier formation, homeostasis, to coordinate formation, shape, Erenewal, and restoration require cells to inte- and function of simple epithelial sheets (Abedin grate different intercellular adhesive cues, cyto- and King 2008; Hulpiau et al. 2013; Miller et al. skeletal dynamics, and signaling in- and output. 2013). This evolution drove the formation of Central to the evolution of multicellular meta- the first intercellular junction, the adherens zoans was the ability to connect adhesion at in- junction (AJ), in which the plasma membrane tercellular junctions with the organization of the served as an organizing platform for adhesive

4These authors contributed equally to this work. 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.a029207

1 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

complexes. Ultimately, these Hatzfeld et al. 2017; Mege and Ishiyama 2017; primitive junctions provided a blueprint for Yap et al. 2017). What is much less appreciated the formation of specialized, spatially and is how cells integrate different junctional me- structurally defined junctional adhesive and/or chanical and signaling activities into a higher barrier-forming complexes along the basal to order network essential to coordinate cell shape apical axis (Rodriguez-Boulan and Macara and positioning with tissue renewal, differenti- 2014), essential for managing the functions of ation, and regeneration. simple epithelia (Fig. 1A). These functions in- In this review, we will highlight the literature clude spatial regulation of signaling, vectorial that uncovers synergy between cadherin adhe- vesicle transport, tissue specific barrier function sive intercellular junctions in tissue architecture and, especially, regulation of cytoskeletal dy- and/or signaling. We will first touch on the evo- namics to control cell and tissue mechanics cru- lutionary significance of cadherin-based junc- cial for tissue morphogenesis, homeostasis, and tions and their molecular components. We will regeneration. then briefly introduce the main lessons on junc- Later in evolution, additional epithelial tional interdependence in simple epithelia and complexity arose in the form of stratified epithe- nonepithelial tissues and discuss recent litera- lia and their appendages. The most visible ex- ture on AJs and desmosomes in the epidermis ample is the epidermis, the first line of defense that show how these cadherin-based junctions against water loss, mechanical insults, and path- integrate adhesive, mechanical, and kinase- ogens in higher vertebrates (Fuchs 2007; Watt transmitted signals to control cell shape and/or 2014). This constantly regenerating barrier bal- differentiation. Finally, using the mammalian ances proliferation in the basal layer with a tight- epidermis, we will propose a model by which a ly controlled differentiation program in which highly synergistic and dynamic intercellular cells move upward while undergoing stepwise junctional network provides a template for or- transcriptional and cell shape changes to form ganizing tissue structure tailored for tissue-spe- the distinct suprabasal layers: the stratum spino- cific functional requirements. sum, stratum granolusum and stratum corneum (Fig. 1B). Distribution of chemical signaling that controls proliferation and differentiation must MAMMALIAN CADHERIN-BASED INTERCELLULAR JUNCTIONS: A BRIEF thus be closely coordinated with the adhesive INTRODUCTION and cytoskeleton machinery that drives the structural cell shape changes associated with Intercellular junctions allow cells to adhere and formation of this stratified physical barrier. communicate with each other while also sepa- What evolutionary strategies were used to rating tissues from the external world or from create such additional tissue complexity re- each other. Organisms also adopted these junc- quired to correctly distribute the chemical and tions as spatially defined signaling platforms mechanical apparatus in a highly patterned and that allowed them to orchestrate cyto-archi- reproducible 3D fashion? To accommodate this tectural changes with signal communication increasing organismal and tissue-specific com- (see, e.g., Chiasson-MacKenzie and McClatchey plexity, junctions and their core components 2017). Specialized mammalian junctions in- diversified from adhesive actin-linked AJs and clude the tight junctions (TJ) that form a para- barrier-promoting tight and septate junctions cellular barrier ion- and size barrier (Van Itallie to channel-forming gap junctions (GJs) and, and Anderson 2014; Balda and Matter 2016) in vertebrates, adhesive intermediate filament and GJ that form small molecule channels to (IF)-linked desmosomes (Green et al. 2010). promote intercellular communication (Nielsen Each of these junctions have been studied exten- et al. 2012). Here, we will focus on cadherin- sively with respect to composition and their ad- based intercellular junctions as a paradigm for hesive, mechanical, signaling or barrier proper- adhesion complexes found in mammalian epi- ties (see, e.g., Braga 2017; Delmar et al. 2017; thelia (Fig. 2).

2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ dacdOln ril.Ct hsatceas article this Cite Article. Online Advanced

ABSimple epithelia Stratified epithelia: epidermis Cornified

Tight junction onSeptember26,2021-PublishedbyColdSpringHarborLaboratoryPress SG1SG2SG3 Dsc1 Pkp1 Pkp3 Dsg4 Adherens junction Intermediate Dsg1 K1/10 Actin filament Granular odSrn abPrpc Biol Perspect Harb Spring Cold

Focal adhesion Vinc vltoayPrpcieo deesJntosadDesmosomes and Junctions Adherens of Perspective Evolutionary E-cad Spinous o:10.1101/cshperspect.a029207 doi: Basal P-cad K5/14 Dsc2 Dsg2 Dsg3/Dsc3 Pkp2

Figure 1. Apicobasolateral polarization of junctions and the cytoskeleton in epithelia. (A) In simple epithelia and (B) in the epidermis as an example of stratified epithelia. The spatial distribution of adherens junction (AJ) and desmosomal proteins across the different layers of the epidermis is also shown. 3 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

AB

Cell membrane

E-cadherin Desmocollin

p120 Plakoglobin -catenin Plakophillins Desmoplakin -catenin

Intermediate F-actin filaments

Figure 2. Schematic representation of core composition of (A) adherens junction and (B) desmosomes.

Adherens Junctions the mechanical strength of AJs (see, for exam- ple, Mege and Ishiyama 2017; Yap et al. 2017). AJs are multiprotein complexes that mechani- For example, force will stabilize the interaction cally couple cell– to the F-actin of the core cadherin–catenin complex with ac- cytoskeleton. In addition to AJs at the lateral tin through a catch bond (Buckley et al. 2014). membrane, simple epithelia have developed a One of the best characterized mechanical AJ specialized AJ, the apically positioned zonula pathway is the regulated recruitment of vinculin adherens (ZA). Members of the classical cad- to the AJs, which relies on a force-dependent herin family of proteins, for example, E-cad- conformational change in α-catenin that un- herin, N-cadherin, VE-cadherin, and P-cad- masks a vinculin-binding site (Leckband and herin, form the calcium-dependent adhesive de Rooij 2014; Ladoux et al. 2015). Several other backbone of AJs that through homophilic and/ mechanosensitive interactions at AJs have re- or heterophilic interactions connect cells (Nies- cently also been identified (Bays et al. 2017; sen et al. 2011). At their cytoplasmic face, this Conway et al. 2017; Hart et al. 2017). Based cadherin subfamily interacts with the armadillo on the magnitude of the cadherin adhesome repeat proteins p120ctn and β-catenin and (Guo et al. 2014), it is likely that many of the through the latter connect via α-catenin with described interactions will be subject to me- the actin cytoskeleton (Fig. 2A). chanical- and/or signaling-dependent regula- Through this core complex, can tion. The challenge will be to identify condi- interact with a range of other cytoskeletal linker tions that reveal the physiological relevance molecules, adaptor proteins, and signaling pro- for many of these interactions. teins, as was shown by both targeted biochem- ical and cell biology analyses as well as recent Desmosomes unbiased proteomic analysis (Padmanabhan et al. 2015). The latter was also referred to as Like AJs, desmosomes are cadherin-based, mul- the cadherin adhesome. Cadherins can not only tiprotein complexes that couple intercellular ad- mechanically sense but also respond to extracel- hesion not to the actin cytoskeleton, but instead, lular and intracellular mechanical signals to to the IF system (Fig. 2B). Desmosomes are modulate actomyosin connections and thus mainly found in epithelia and in the heart of

4 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

vertebrates. The desmosomal cadherin subfam- tion, and immune homeostasis. Null mutations ily consists of (in humans, Dsg1–4) in the cadherin Dsg1 or DP result in a metabolic and desmocollins (in humans, Dsc1–3) that wasting syndrome further characterized by in- show tissue- and differentiation-specific expres- flammation (Samuelov et al. 2013; McAleer et al. sion patterns. How desmosomal cadherins pro- 2015). These keratinocytes also show increased mote adhesion is still not well understood but it expression of inflammatory mediators, thus has been reported that both homophilic and highlighting the potential relevance of these heterophilic interactions between desmogleins other functions. Albeit, these phenotypes could and desmocollins can occur. The extent to also be secondary effects resulting from dis- which one or the other functions in vivo is not turbed epithelial barrier function as a conse- known. The desmosomal cadherin cytoplasmic quence of impaired intercellular cohesion. It is domains bind to the armadillo protein plakoglo- thus essential to consider the desmosome as an bin, a β-catenin homolog, and to the plakophilin integrator of mechanics and signaling that coor- (PKP) family of armadillo proteins. These arma- dinates tissue structure with function when ex- dillo linker proteins then interact with the IF- amining the underlying causes of phenotypes binding protein desmoplakin (DP), thus con- observed in desmosomal diseases. necting desmosomes to the IF cytoskeleton Testifying to the notion that AJs and desmo- (Fig. 2B). For further details on desmosomal somes functionally integrate adhesive, cytoskel- composition and function, we refer to several etal, and signaling responses, it is important to outstanding reviews (Delva et al. 2009; Thoma- note that several mammalian tissues/organs son et al. 2010; Broussard et al. 2015). Unlike have molecularly distinct or mixed junctions AJs, desmosomes can adopt a hyperadhesive crucial for tissue function. For example, the ma- state that is independent of calcium and for ex- jority of intercellular junctions in cardiomyo- ample, controlled through protein kinase C cytes of higher vertebrates are hybrid junctions (PKC)-dependent posttranslational modifica- (Franke et al. 2009; Vite and Radice 2014) that tions in desmosomal components (Garrod and contain components of both AJs and desmo- Tabernero 2014; Hatzfeld et al. 2017). somes. Similarly, AJs in vascular endothelial As mutations in different desmosomal com- cells contain desmosomal components like pla- ponents result in skin blistering and sudden car- koglobin and DP and can connect to the IF cy- diac arrest syndromes (reviewed in Samuelov toskeleton as well (Franke et al. 2009). Thus, in and Sprecher 2015), desmosomes are considered these tissues, the AJ and desmosome systems essential for providing mechanical strength to integrate their function also on a structural level. tissues. Studies indicate that the desmosome/IF system, like the AJ/actin system, is not only im- portant for withstanding mechanical stress but EVOLUTIONARY ORIGINS OF CADHERIN- DEPENDENT ADHESION AND also instructive in the establishment of the in- CYTOSKELETAL ORGANIZATION herent mechanical properties of cells. Keratino- cytes deficient for all keratins revealed that ker- The ability of cells to form multicellular clusters atins are key determinants of cell stiffness to with physical behaviors mimicking a fluid that control their migratory behavior (Ramms et al. support intercellular rearrangements is consid- 2013; Seltmann et al. 2013). A very recent study ered a major transition in evolution that drove showed that the IF-binding domain within DP the appearance of metazoa (King 2004; New- controls intercellular tension and cell stiffness, man 2016). Acquisition of these material prop- which in part are mediated through regulation of erties that enable cellular populations to under- the actin cytoskeleton (Broussard et al. 2017). go morphogenesis requires coordination and/or It is becoming increasingly clear that des- coupling of intercellular adhesion and cytoskel- mosomal components as well as IF, and thus etal dynamics to control cell–cell rearrange- likely desmosomes as a whole, also serve a range ments and cell shape of the first primitive epi- of other purposes related to growth, differentia- thelia. The appearance of classical cadherins in

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 5 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

one of the earliest metazoa, sponges, that medi- The reasons for this relatively rapid expan- ate intercellular adhesion and through their sion of cadherins in one of the closest relatives of newly evolved cytoplasmic domain engaged cat- metazoa as well as their function are not entirely enins to control and the cytoskele- clear. The colony-forming variants of choano- ton (Oda and Takeichi 2011), was thus a crucial flaggelates form cell–cell contacts through cyto- step for such a transition. Interestingly, the two plasmic bridges. One theory is that these cad- molecular groups necessary for this transition, herins are important for capture of bacterial cadherins and cytoskeletal linker/adaptor mol- prey and subsequent feeding (Nichols et al. ecules, seem in part to have evolved indepen- 2012). Additional evidence for such a role comes dently from each other. from the observation that both sponges and choanoflaggelates have a motile flagellum used to generate water flow to capture prey on a sur- Evolution of Classical and Desmosomal rounding microvillar collar where these bacteria Cadherin Adhesion Receptors are phagocytosed (Alegado and King 2014). Members of the cadherin superfamily are typi- One of the cadherins of Meloe brevicollis has cally multidomain proteins that have evolved been shown to localize to this collar (Abedin through duplication, divergence, as well as shuf- and King 2010; Pizarro-Cerda et al. 2012). In fling of domains between different families (Oda agreement with a potential evolutionary con- and Takeichi 2011; Hulpiau et al. 2013). The served function in bacterial recognition and in- recent identification of a single cadherin in Cap- ternalization, human E-cadherin serves as a re- saspora owczarzaki, the unicellular outgroup of ceptor for listeria that is necessary for its choanoflaggelates and metazoan lineages, indi- internalization (Pizarro-Cerda et al. 2012). cates that cadherins predate the divergence of The last evolutionary addition to the cad- C. owczarzaki, choanoflagelate, and metazoan herin superfamily is the desmosomal cadherin lineages (Nichols et al. 2012). Interestingly, the family, found only in vertrebrates (Broussard two species of choanoflaggelates thus far ana- et al. 2015). What drove the evolution of desmo- lyzed both have >20 types of cadherin molecules somal cadherins is not entirely clear. It is note- that contain cytoplasmic domains unrelated to worthy, nevertheless, that their appearance co- classical cadherins. These can be further classi- incided with further evolutionary developments fied in at least three families, two of which are such as the expansion of epithelial tissue com- thus far only found in sponges and choanoflag- plexity, including stratified epithelia, primitive gelates, the lefftry and cohesion families. The hearts consisting of multiple chambers that lefftry family is characterized by a laminin ami- were connected to an outward flow (Olson no-terminal domain, four epidermal growth 2006), and the arrival of a more complex im- factor receptor (EGFR) repeats, and a furin re- mune system including the adaptive immune peat; one or two extracellular fibronectin 3 re- system (Cooper and Alder 2006). peats located close to the transmembrane do- Interestingly, the extracellular domains of main; as well as a cytoplasmic inactive tyrosine vertebrate classical and desmosomocal cadher- phosphatase domain. The sponge Oscarella car- ins no longer combine cadherin extracellular mela also contains a lefftry family member but (EC) domains with other extracellular matrix with an active PTP domain. The cohesion family (ECM)-like and/or signaling-like repeats unlike, contains a bacterial/archaeal cohesin domain. for example, mammalian CelsR/Flamingo or Fat Hedgling, the third family, is found in choano- nonclassical cadherin family members or Dro- flaggelates, sponges, and cnidarians but is absent sophila classical cadherins (Oda and Takeichi from bilaterians. This family is characterized by 2011; Hulpiau et al. 2013). The increasingly an amino-terminal hedgehog domain connect- complex morphogenetic movements and/or in- ed to a Von Willebrand A domain and many creasing number of epithelial tissues later in members contain several epidermal growth fac- metazoan evolution might thus have required tor receptor (EGF) repeats (Nichols et al. 2012). structural separation of cadherin repeat function

6 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

from signaling and ECM-like domain function cadherin adhesome orthologs predate metazo- to allow diversification of signaling and adhe- ans (Murray and Zaidel-Bar 2014); albeit, it is sion. Alternatively, domains other than the cad- not entirely clear whether some of these were herin repeat domain may have been lost in clas- able to interact with premeatozan cadherin cy- sical and desmosomal cadherins, as their toplasmic domains. The best evidence for ini- presence may have hampered increasingly dy- tially independent evolution of cadherins and namic adhesive interactions required to drive the AJ complex members, α- and β-, expanding epithelial complexity. This selective comes from the amoeba Dictyostelium discoi- pressure might have been less strong in other dum. This social amoeba does not have any cad- classical cadherin super family members that herin-like molecules but contains an α-catenin/ are involved in intercellular interactions but vinculin homolog and a β-catenin-like armadil- not strong adhesion itself. lo repeat protein, Aardvark, which can interact with mammalian α-catenin as well, thus indi- cating functional conservation. Importantly, Cadherin–Cytoskeletal Linker Proteins these two proteins are essential for actin organi- Both AJ and desmosomal cadherins directly in- zation and the formation of an epithelial-like teract with so-called Armadillo (Arm) repeat cell sheet that surrounds the tip of the stalk proteins, which contain a domain first named during the multicellular stage of Dictyostelium after the Drosphila β-catenin homolog armadil- (Grimson et al. 2000; Dickinson et al. 2011, lo. This Arm repeat is an evolutionarily old 2012b). Although in Dictyostelium, α- and β- motif that is also found in a range of other pro- catenin are associated with junctions that recruit teins, such as importins (Gul et al. 2017). Clas- actin and ultrastructurally resemble AJs, it is not sical cadherins interact with β-catenin/plako- clear how these proteins associate with the cor- globin and p120-catenin members of the Arm tex. Moreover, unlike metazoa, the formation of repeat family, whereas desmosomal cadherins these AJ-like structures occurs independently of associate with plakoglobin and the plakophilin either α-orβ-catenin (Dickinson et al. 2012a). It family that belong to the p120 super family. will thus be interesting to determine whether in These interactions are essential to link these social amoebae the function of α- and β-catenin adhesive systems to their respective cytoskele- in cytoskeletal and epithelial organization was tons via either α-catenin (classical cadherins) independent of adhesion or requires interaction or DP, respectively (Green et al. 2010). As with one of the adhesion molecules found in well, they regulate cell surface stability and/or Dictyostelium, such as the calcium-dependent, control of signaling pathways crucial for cyto- noncadherin Cad molecules or the IgG-like skeletal dynamics (Kourtidis et al. 2013; adhesion receptors (Siu et al. 2004). Thus, AJ Hatzfeld et al. 2014). Interestingly, albeit outside cytoplasmic plaque proteins were one driving the scope of this review, all these cadherin-in- evolutionary force for promoting cell sheet for- teracting Arm repeat proteins can also translo- mation and epithelial polarization. cate into the nucleus and interact with transcrip- In contrast, the p120 super family of pro- tion factors (e.g., p120) or the translational teins may have coevolved with the cadherins. machinery (e.g., Pkp1) (reviewed in McCrea Poriferans (sponges), the most-simple metazo- and Gottardi 2016). The best characterized ex- ans, already contain both a classical cadherin ample is β-catenin that, next to its crucial func- and a p120ctn armadillo repeat family member tion in adhesion, is a central player of the Wnt that is most closely related to vertebrate δ-cate- pathway, which evolved also around the time of nin (Carnahan et al. 2010). Unlike in verte- the first metazoa (Pires-daSilva and Sommer brates, in which p120ctn is essential for cadherin 2003). function, in lower organisms such as C. elegans The initial evolution of these linker mole- and Drosophila, p120ctn binding is not crucial cules may in part have occurred independently but serves to modulate cadherin complex func- from the cadherins. Approximately 70% of the tion (Myster et al. 2003; Pacquelet et al. 2003;

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 7 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

Hardin 2015; Bulgakova and Brown 2016). (PRDs) that associate with IFs to confer struc- p120ctn super family members control cadherin tural integrity and resilience to tissues (Leung cell surface stability and regulate the activity et al. 2002). Plakins, first observed in chordates, of the Rho family of small GTPases (Kourtidis most likely arose from the spectraplakin family, et al. 2013; Hatzfeld et al. 2014). These GTPases which was first found in sponges and character- are key regulators of actomyosin dynamics, ized by their ability to directly engage different junctions, and cell shape (Garcia-Cattaneo and cytoskeletal systems, either in a single protein or Braga 2013). Thus, very early on, classical cad- through expression of splice variants (Huels- herins may have gained the ability to not only mann and Brown 2014). In C. elegans, one splice interact with the cytoskeleton but through inter- variant of a spectraplakin, vab10a, is essential to actions with p120ctn members dynamically link IFs to in epidermal cells, regulate surface tension and thus intercellular an adhesive structure that connects muscle to rearrangements. the cuticle (Bosher et al. 2003), thus sharing a In vertebrates, the p120ctn family expanded key feature with DP. DP itself likely coevolved to 7 members (Carnahan et al. 2010), likely to with desmosmal cadherins in vertebrates. adapt to the increasingly different morphoge- Taken together, then, there were two deci- netic demands in tissue complexity as reflected sive evolutionary developments for intercellular in the expansion of vertebrate classical and des- junctional cadherin complexes: mosomal cadherins. The ubiquitously expressed 1. Convergence of the initially independent evo- p120ctn may have coevolved with nonneuronal lution of cadherin adhesive domains and the cadherins whereas δ-catenin became mostly re- actin-engaging α-, β-catenin module, with stricted to neuronal lineages, and may have the later appearing classical cadherin cyto- further evolved with predominantly neuronally plasmic domain enabled organisms to couple expressed cadherins (N- and R-cadherin). cell adhesion to cortical tension. The simulta- Moreover, PKP1–3, and the β-catenin homolog neous arrival of the first p120-like molecule plakoglobin, are only found in vertebrates (Zhao likelyallowed these simple early metazoans to et al. 2011) thus coinciding with the arrival of dynamically and mechanically control the desmosomal cadherins on the evolutionary cadherin–cytoskeletal link to drive intercellu- stage. This similar timing indicates a mutual lar morphogenetic rearrangements. It is dependence of function early during vertebrate worth noting that keysignaling receptors nec- development. PKP proteins are crucial in re- essary for intercellular communication and cruiting DP, and thus IFs, to desmosomes. In fate determination, such Wnt and receptor addition, these proteins in part may have similar tyrosine kinases also appeared at the same functions as p120 as these proteins can regulate time with the classical cadherin/catenin com- actin and Rho family activity (Hatzfeld et al. plex (Pires-daSilva and Sommer 2003). It may 2014), thus allowing the newly arrived desmo- thus well be possible that signaling and adhe- some to communicate with the evolutionary sion-dependent mechanics were integrated older AJ. early during metazoan evolution. DP, the molecule that links desmosomal cadherins to IFs through plakophilins and pla- 2. The simultaneous evolutionary diversifica- koglobin, is a member of the plakin family of tion of cadherins, p120, and β-catenin arm cytoskeletal linkers. Proteins in this family are families, and plakin family into desmosomal characterized by amino- and carboxy-terminal cadherins, PKPs, plakoglobin, and DP, re- globular domains flanking a central a-helical spectively, in vertrebrates enabled the forma- coiled-coil rod. The amino-terminal plakin do- tion of desmosomes to engage IFs. These main comprises a series of spectrin repeats and a junctions thus provide mechanical stability Src-homogy 3 domain involved in junctional and resilience likely necessary to accommo- targeting and the carboxy-terminal domain date more complex tissue structures such as a comprises a series of plakin repeat domains multichambered heart, or stratifying epithe-

8 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

lia. These early vertebrates also faced new desmosomal cadherins Dsg2 (Eshkind et al. challenges in the terrestrial environment, 2002) and Dsc3 (Den et al. 2006) or the cyto- such as sunlight, and exposure to different plasmic linker proteins DP (Gallicano et al. pathogens; desmosomes may then have con- 1998), are early embryonic lethal in mice. Sim- tributed to newly acquired epidermal barrier ilarly, loss of zebrafish Dsc and Dsg interferes functions including integration of the innate with epiboly (Goonesinghe et al. 2012), further immune with the adaptive immune system revealing important roles for these proteins in that arose at the same time. early morphogenetic cellular rearrangements. The majority of these phenotypes are associated with impaired desmosomal adhesive or IF link- INTERPLAY BETWEEN JUNCTIONS: age function, leading to increased tissue fragility. LESSONS FROM EARLY DEVELOPMENT, The role of desmosomal proteins in the regula- SIMPLE EPITHELIA, AND HEART tion of other junctions or other functions apart Tissue morphogenesis, homeostasis, and regen- from providing mechanical stability have not eration require coordinated cellular rearrange- been well studied in these early lethal mutants. ments. Early studies identified AJs as key regu- Mouse knockouts for those desmosomal pro- lators of epithelial structure and intercellular teins that did not result in early embryonic le- junction formation. Complete loss of E-cad- thality (e.g., Dsg3, Dsc1, PG, PKP1) resulted in herin, β-catenin, or α-catenin, in Drosophila, impaired heart development and/or skin blister- C. elegans, Xenopus, Zebrafish, or mice results ing (McCauley and Wehrens 2009; Ganeshan in early embryonic lethality with severe defects et al. 2010), thus providing direct evidence that in epithelial integrity, junction formation, and the observed mutations in the human gene polarity (reviewed in Harris and Tepass 2010). counterparts or the auto-antibodies against des- Moreover, in simple epithelial cell culture mod- mosomal cadherins are indeed causal for the els, antibodies to E-cadherin interfere not only associated diseases (Samuelov and Sprecher with AJs but also desmosome, GJ, and TJ for- 2015). mation (Gumbiner et al. 1988). Although in sev- The role of desmosomal proteins in simple eral simple epithelia (e.g., liver, mammary, or epithelia has only been marginally explored. thyroid gland), loss of E-cadherin did not obvi- Surprisingly, inactivation of DP in the mouse ously affect intercellular junctions (Boggetti and intestinal did not obviously affect Niessen 2012), loss of E-cadherin in intestinal AJs, TJs, or intercellular adhesion, resulting in epithelial cells impaired junctions and barrier viable mice, suggesting that desmosomes are function (Schneider et al. 2010; Bondow et al. dispensable in this tissue (Sumigray and Lechler 2012). In keratinocytes, loss of E-cadherin is 2012). Instead, microvilli structure is shortened, sufficient to interfere with TJs, whereas only thus providing further in vivo evidence that des- combined loss of both classical cadherins, E- mosomes affect actin-based structures, similar and P-cadherin, prevented formation of desmo- to what has been observed in keratinocytes (Va- somes in these cells (Michels et al. 2009). Clas- sioukhin et al. 2001b). In contrast, CRISPR/Cas- sical cadherin-dependent AJs may thus serve as mediated inactivation of Dsc2 and Dsg2 in co- master regulators to initiate the formation of lon carcinoma DLD1 cells caused intercellular intercellular junctions in simple epithelia. In fragility accompanied by the absence of desmo- agreement, cell culture and organismal models somes but not AJs and TJs (Fujiwara et al. 2015). reveal that AJ formation precedes the assembly Together, these data thus suggest that in simple of desmosomes, GJs, and TJs (Fleming et al. epithelia desmosomes do not obviously regulate 1994; Vasioukhin et al. 2001b). the formation of other junctions. Having said Even though known human diseases arising that, in the future it will be essential to examine from mutations related to desmosomal proteins the function of different desmosomal proteins (see below) suggest that desmosomes are only in other simple epithelia as well as challenge essential in heart and skin, inactivation of the desmosomal-specific knockout mice that may

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 9 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

unravel stress-associated functions for desmo- basal to apical axis of the entire tissue through somes in intercellular junctional rearrangements as yet poorly understood mechanisms (Fig. 1B; and tissue regeneration. Muroyama and Lechler 2012; Tellkamp et al. Interestingly, recent studies in the heart have 2014). Below, we will briefly introduce the epi- revealed an important role for desmosomes in dermis and address the contribution of AJs and controlling GJ function in addition to their desmosomes in epidermal polarity, structure, mechanical contribution at the intercalated and function. disc. Cardiomyocytes are electrically coupled The self-renewing epidermis forms the out- through GJs, essential for cardiac rhythm and ermost skin structural and innate immune bar- contraction. Mice with a cardiomyocyte-specific rier that protects from dehydration and external deletion of DP show loss of -40 and challenges, such as UV and microbes (Blanpain -43, the molecular building blocks of the GJ and Fuchs 2009). This tissue is a remarkable channels, accompanied by right ventricular con- example of exquisite and robust control of duction defects, leading to AC (Lyon et al. 2014). continuous, dynamic cellular rearrangements Similarly, cardiac loss of Dsg2 or cardiac expres- and cell-shape changes while cells are en route sion of an adhesion-defective Dsg2 mutant re- from the basal to the stratum corneum layer, all sults in AC and an altered connexin-43 distri- while retaining an intact barrier. To maintain bution, which was linked to the adhesive homeostasis or to restore injury without inflict- function of Dsg2 (Kant et al. 2015). Mutations ing disease thus requires integration of signals in human DP that are linked to AC also result in that regulate barrier architecture with those that junctional loss of connexin-43 (Samuelov and govern innate immunity and metabolic activity. Sprecher 2015). The molecular mechanism re- Improper development or disturbed mainte- sponsible remains to be elucidated. DP muta- nance of the epidermal barrier results in a range tions disrupt binding to the microtubule-asso- of skin diseases including common inflamma- ciated protein end-binding1 (EB1), which tory skin diseases, such as atopic dermatitis and regulates trafficking of connexin-43 to GJs (Patel psoriasis, impaired healing, and skin cancer et al. 2014). Desmosomes may thus stabilize mi- (Kubo et al. 2012). crotubules (MTs) through EB1, providing a spa- The process of stratification that generates tial cue for membrane targeting of connexin-43. this multilayered epithelial barrier requires a Together, these results provide a potential mo- spatiotemporally orchestrated set of transcrip- lecular explanation on how desmosomes regu- tional and architectural changes that result in late GJ stability. complete remodeling of epidermal cell shape and function (Candi et al. 2005; Simpson et al. 2011). During this process, cell architectural fea- POLARITY AND JUNCTIONS IN STRATIFIED tures such as junctions and cytoskeletal proteins EPITHELIA are polarized across layers along the basal to Many of the insights regarding junctional co- apical axis of the stratified epithelium. Perhaps, operativity in the formation, maintenance, and the best example for epidermal tissue polariza- restoration of epithelial tissues come from tion is the restricted localization of the TJs to the studies performed on the epidermis, a stratified second granular layer (SG2) of the epidermis epithelium. Unlike simple epithelia, stratified (Fig. 1B). Recent whole-mount analysis of new- epithelia, also including epithelia of the esoph- born and adult mouse and human epidermis agus, urinary tract, and oral cavity, constitute showed that TJs localize apically within these two or more layers. These additional layers cells (Yokouchi et al. 2016; Rübsam et al. in essentially make it impossible to establish api- press). These TJs are essential to prevent unnec- cobasolateral polarity within one layer, as ob- essary water loss from the organism (Furuse served in simple epithelia. Instead, stratified et al. 2002; Tunggal et al. 2005), an essential epithelia establish polarity of cytoskeletal orga- feature for terrestrial life. Molecularly, only oc- nization and intercellular junctions along the cludin and -4 are specifically found in

10 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

the stratum granulosum, whereas ZO-1 and tion indicate that the different layers adopt ar- claudin-1 are present in all cell layers, suggesting chitecturally and mechanically different states other functions outside of TJs for these proteins. likely essential to drive layer-dependent transi- Although desmosomes and AJs are present in all tions and functions. layers, the molecular components of these junc- tions differ between the specific layers; basal lay- ers express E- and P-cadherin as well as Dsg3. JUNCTIONS COORDINATE EPIDERMAL ORGANIZATION, GROWTH, AND On initiation of differentiation and movement DIFFERENTIATION into the first suprabasal layer, P-cadherin, Dsg2, Dsg3, and Dsc3 are down-regulated whereas Evidence indicating that junctional organization Dsg1 is up-regulated with further increasing ex- of the cytoskeleton is important for tissue orga- pression in suprabasal layers (Fig. 1B). Although nization and mechanics comes from in vivo epi- E-cadherin containing AJs are found in all lay- dermal loss-of-function mouse models, 3D or- ers, vinculin, a mechano-sensitive component ganotypic culture models, and from human skin of AJs, is only highly enriched in the SG2 layer, diseases associated with mutations in, or anti- in which TJs are formed (Rübsam et al. in press). bodies against, junctional components. For ex- Intriguingly, Dsg4 expression seems to be con- ample, auto-antibodies against Dsg3 or Dsg1 re- fined only to the SG1 and the SC. Similarly, sult in skin blistering diseases with separation PKP1 and 3 show a counter-gradient across lay- either between basal and suprabasal layer ers, whereas plakoglobin, DP, and PKP2 are (Dsg1 and Dsg3) or upper suprabasal layers found in all layers. This layer-specific expression (Dsg1). Mutations in desmosomal cadherins, and recruitment to junctions is functionally im- plakoglobin, PKP1, PKP3, or DP result in a range portant, as inappropriate in vivo expression of of human diseases characterized by skin blister- desmosomal cadherins, E-cadherin, or claudin- ing and/or AC, the severity of which depends 6 interferes with epidermal morphogenesis and both on the specific gene and type of mutation barrier function (Elias et al. 2001; Henkler et al. (Lai-Cheong et al. 2007; Samuelov and Sprecher 2001; Merritt et al. 2002; Turksen and Troy 2015). As noted earlier, these human phenotypes 2002; Jamora et al. 2003). by and large have been confirmed by the respec- The cytoskeletal networks also show differ- tive knockout mice. In contrast, P-cadherin mu- ent layer-dependent configurations: Although tations in humans are associated with a mild ec- MTs form a centrosomal network in the basal todermal dysplasia syndrome predominantly layer, they concentrate at cell–cell junctions in characterized by hair loss (Lai-Cheong et al. suprabasal layers. Interestingly, this suprabasal 2007), whereas mice deficient for P-cadherin redistribution of MTs depends on recruitment showed no obvious skin phenotype (Tinkle of centrosomal proteins to DP at the junctional et al. 2008). Interestingly, combined loss of P- area (Sumigray et al. 2011). The keratin filament cadherin and desmoglein-3 results in a much system consists of pairs of type I and type II stronger oral blistering phenotype than loss of keratins (see also, Hatzfeld et al. 2017). The basal desmoglein 3 alone (Lenox et al. 2000), showing layer expresses K5/K14 whereas suprabasal lay- that synergy between AJs and desmosomes also ers express K1/K10-based networks. Thus, des- occurs on the tissue level. As anticipated, loss of mosomes are connected to keratin networks of E-cadherin alone or in combination with P-cad- different composition and likely varying me- herin or loss of α-catenin disturbs F-actin orga- chanical resilience in basal versus suprabasal nization (Rübsam et al. in press; Vasioukhin et al. layers. Finally, F-actin, like MTs, is differentially 2001a; Tinkle et al. 2008). In agreement E-cad- organized across layers with the highest cortical herin both intercellular and cortical tension in F-actin organization in the granular layer (Fig. keratinocyte cell sheets. However, it was unex- 1B) (Rübsam et al. in press). Together, these pected that loss of E-cadherin disturbed the po- highly specific and differential distributions of larized tissue organization of F-actin, resulting in adhesion molecules and cytoskeletal organiza- increased cortical F-actin organization in lower

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 11 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

layers (Rübsam et al. in press), suggesting that in a decrease in cortical actin remodeling after cad- these lower layers E-cadherin actively restricts herin engagement (Godsel et al. 2010). More- cortical F-actin organization. over, PKP2 loss, at least in part through RhoA, decreases cell spreading, increases stress fiber formation, and induces more stable focal adhe- Desmosomal Regulation of Actin-Based sions, thus reducing cell migration rates (Godsel Junctions et al. 2010; Koetsier et al. 2014). Perturbations of desmosomal components not only disturb mechanical integrity directly linked Junctional Control of SG2 Tight Junctional to desmosomal/keratin-mediated dysfunction Barrier Formation but often also result in alterations in cell behav- iors known to be regulated by the actin cytoskel- Unlike simple epithelia, there is clear evidence eton, such as cell migration. For example, epi- for a role of desmosomes in the formation and dermal inactivation of DP is perinatal lethal function of epidermal TJs. For example, ectopic because of mechanically stress-induced loss of expression of Dsg3 in the upper layers of mouse epidermal integrity (Vasioukhin et al. 2001b). epidermis results in neonatal lethality as a result Interestingly, DP null keratinocytes not only of increased transepidermal water loss (Elias have reduced desmosomes that no longer are et al. 2001), suggesting impairment of TJs, al- linked to the keratin network but also reduced though the SC barrier is also altered. In contrast, AJs. These keratinocytes show increased myosin inactivation of PKP1 in mouse epidermis did contractility and are unable to properly reorga- not functionally alter the outside-in SC barrier nize the actin cytoskeleton (Vasioukhin et al. but did impair TJ barrier function, likely as a 2001b; Sumigray et al. 2014). This regulation consequence of impaired intercellular junction likely requires binding of DP to IFs as deletion formation. Moreover, DP was shown to control of this interaction domain altered actin-depen- claudin expression (Sumigray et al. 2014). Thus, dent intercellular tension (Broussard et al. there are many indications that desmosomes not 2017). only affect AJs but also TJs, which perhaps is not Vice versa, either combined loss of the main that surprising taken their role in regulating ac- epidermal classical cadherins, E- and P-cad- tin dynamics. herin, or loss of α-catenin in keratinocytes is The mechanisms that confine TJs to the SG2 perinatal lethal and strongly impairs both AJ layer are mostly unknown. Very recently, it was and desmosome assembly (Vasioukhin et al. shown that E-cadherin is essential to restrict TJs 2001a; Tinkle et al. 2008; Michels et al. 2009). to the SG2. Epidermal loss of E-cadherin or β- Along these lines, loss of Dsg1 in 3D human catenin resulted in breaches in the SG2 TJ barrier epidermal cultures also results in increased cell (Tunggal et al. 2005; Ray et al. 2013). Similar to size and variability of cell shape (Getsios et al. increased cortical F-actin organization, prema- 2009), characteristics linked to actin organiza- ture spot-like TJ-like structures were observed in tion. Together, these results indicate that AJs lower layers up on loss of E-cadherin. One po- and desmosomes act synergistically to promote tential mechanism is that E-cadherin integrates epidermal integrity while at the same enabling junctional tension and EGFR activity to inhibit individual cell shape changes during their trav- premature TJ complex formation in lower layers erse through the different layers. while promoting increased tension and TJ stabil- There are multiple examples linking desmo- ity in the SG2 (Fig. 4B) (Rübsam et al. in press). somes with upstream and downstream regula- tors of actin organization (Green et al. 2010; Epidermal Junctions in Signaling and Hatzfeld et al. 2014). One involves PKP2. In Differentiation both squamous cell lines and atrial cardiomyo- cytes, decreased PKP2 levels result in a failure of Recent evidence suggests that AJs and desmo- active RhoA to localize to cell–cell contacts and somes not only guarantee cohesion and barrier

12 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

function but also actively coordinate signaling loops, as EGFR signaling promotes the endo- and the differentiation process itself. Dsg1 pro- cytic turnover of Dsg2 (Klessner et al. 2009). motes keratinocyte differentiation by suppress- As desmosomes and AJs control Rho and ing EGFR and mitogen-activated protein kinase actin dynamics, this potentially provides addi- (MAPK) signaling cascades (Getsios et al. 2009). tional pathways to allow these junctions to inte- Instead, the Dsg1-mediated effects on differen- grate the regulation of cell shape and balanced tiation require its interaction with the cytosolic renewal versus differentiation in the epidermis. protein Erbin (ErbB2 interacting protein), Rho-family dependent modulation of the actin which results in the disruption of Ras–Raf com- cytoskeleton can regulate cell behaviors through plexes mediated by the scaffolding protein transcription. The polymerization of F-actin, SHOC2 (Harmon et al. 2013). Interestingly, downstream from Rho signaling, drives the this mechanism also does not require the adhe- transcriptional coactivator MAL into the nucle- sive ectodomain of Dsg1. In addition to regulat- us where it aids in serum response factor (SRF)- ing differentiation through EGFR-related sig- dependent transcription (Posern and Treisman naling, Dsg1 has been shown to be important 2006), thus linking actin dynamics to differen- for the entry of keratinocytes into the terminal tiation. In keratinocytes, loss-of-function of the differentiation program through the tyrosine ki- RhoA guanine nucleotide exchange factor nase EphA2 (Lin et al. 2010). (GEF) breakpoint cluster region (Bcr) has been Other desmosomal components also have shown to reduce RhoA activity and inhibit an been shown to regulate/be regulated by tyrosine SRF-mediated, prodifferentiation phenotype, kinase signaling. For example, PKP2, which is and restoring the levels of Dsg1 rescues the widely expressed in simple epithelia and con- Bcr-induced differentiation defects (Dubash centrated in the lower layers of stratified epithe- et al. 2013). Along similar lines, increasing evi- lia, is a positive regulator of EGFR activation. dence indicates that AJs control Yap signaling Loss of PKP2 disrupts EGFR signaling and leads through α-catenin mechanical signals to bal- to decreased cancer cell migration and prolifer- ance growth and differentiation (Schlegelmilch ation (Arimoto et al. 2014). PKP3 is present in et al. 2011; Zhang et al. 2011) and that defective most simple epithelia and is expressed more uni- cell adhesion results in unrestricted epidermal formly in stratified epithelia like the epidermis. Yap signaling that promotes epidermal over- PKP3 is phosphorylated in response to pemphi- growth (Li et al. 2016; Walko et al. 2017). gus IgG, resulting in dissociation from Dsg3 and destabilization of adhesion. These effects are di- minished on suppression of Src activity (Cirillo INTEGRATIVE MODELS FOR AJS AND DESMOSOMES MECHANICS et al. 2014). Finally, uniformly expressed plako- AND SIGNALING globin has been shown in both keratinocytes and prostate cancer cell lines to inhibit cell motility As discussed, AJs and desmosomes not only through a mechanism that involves the regula- bidirectionally control their formation and ac- tion of Src and the extracellular matrix (Todo- tivity but also determine cell shape and differ- rovic et al. 2010; Franzen et al. 2012). entiation status. However, whether differentia- Aberrant expression of Dsg2 in the upper tion-dependent signals and control of cell and layers of the epidermis promotes a hyper- tissue architecture are integrated at the level of proliferative phenotype through growth factor desmosomes and AJs is less clear. Using the signaling cascades that stimulate both cell pro- transition from the basal to the first suprabasal liferation and survival (Brennan et al. 2007). layer, as well as the transition from SG1 to SG2 In addition, reduced expression of Dsg2 in in- as two examples, we now propose two models testinal epithelia suppresses cell proliferation for how AJs and desmosomes cooperate to inte- through a reduction in EGFR signaling (Kame- grate mechanical and chemical signals to ro- kura et al. 2014). Moreover, there are potential bustly promote the transition into a new layer growth factor-mediated signaling feedback with different mechanical properties and differ-

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 13 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

entiation status (Figs. 3, 4). These models are essary to establish a mechanical boundary be- based on developmental models in which differ- tween basal and suprabasal cells. ences in tissue surface tension trigger cell sorting In the case of a delaminating cell (Fig. 4A), and cell shape changes to drive tissue organiza- we propose that a combined loss of cell–matrix tion (Amack and Manning 2012; Heisenberg contacts and dynamic AJ and desmosomal re- and Bellaiche 2013). arrangements locally reduce cortical tension compared with its neighbors driving initial up- ward movement and initiation of differentia- Basal to Suprabasal Transition tion. During this process, the basal cadherins At present, two different mechanisms have been P-cadherin and Dsg3 are transcriptionally proposed to explain how basal cells become down-regulated whereas E-cadherin and Dsg1 suprabasal cells: (1) through asymmetric divi- expression increase in the delaminating cell, ac- sion coupled to perpendicular spindle orienta- companied by a switch from a K5/K14 network tions that position one of the daughters more to a K1/K10 network, allowing the delaminating suprabasally; and (2) through delamination by cell to more specifically interact with the supra- loosening cell–matrix contacts. It is well estab- basal cells, which express E-cadherin and a lished that changes in cortical tension transmit- higher Dsg1:Dsg3 ratio. Strengthening desmo- ted through cadherins drive cell–cell segregation somal interactions through DP engagement of and cell sorting to establish tissue boundaries. K1/10 will not only provide more resilience but We propose that initial local remodeling of basal also strengthen E-cadherin AJs (Broussard et al. intercellular junctions alters cortical tension. 2017), thus increasing junctional tension and This local tension disequilibrium is necessary resulting in integration of the delaminating cell to drive (in case of delamination) and/or pro- into the first suprabasal layer (Fig. 3). mote upward movement (in both cases). Subse- In the case of an asymmetric mitotic spindle, quent differentiation-induced changes in key AJ during late anaphase/telophase the future upper and desmosomal adhesion molecules further re- daughter cell membranes are positioned away inforce these intercellular rearrangements nec- from the basement membrane and will start to

Basal to suprabasal transition Delamination P/E-cad adherens junction Mechanical boundary E-cad adherens junction

Basal Actin Matrix attachments Asymmetric cell division Basal desmosome Mechanical Suprabasal desmosome boundary Basal intermediate

Basal filament Differentiated intermediate filament AJ/DSM remodeling AJ switch Junctional tension cortical tension DSM/IF switch Cell-cell adhesion Sorting out into Differentiation suprabasal layer

Figure 3. Cadherin-based junctions in basal to suprabasal transition. Proposed model how dynamic reorgani- zation of adherens junctions (AJs) and desmosomes control tension and differentiation to promote the basal to spinous layer transition either when cells delaminate or up on asymmetric cell division resulting in the formation of a mechanical tissue boundary that allows the tissue to couple cell position and differentiation status.

14 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

A Transition of SG3 to barrier forming SG2

Lack of apical Desmosomes only adherens TJ formation junctions: no High AJ tension apical actin SG2

Granular attached Low AJ tension SG2 SG3 SG1

Desmosome Tight junction Adherens junction AJ-anchored cotical actin Fully attached cortical actin

SG3

B Feedback between junctions, signaling and tension to restrict TJs to SG2 Basal Spinous Granular Cornified

Tight junction SG1 Adherens junction TJ stability SG3 SG2 Actin ? Matrix attachments AJ-tension high EGFR DSM/IF Activity Basal desmosome Asymmetry? TJ stability Suprabasal ? desmosome AJ-tension low EGFR Basal intermediate DSM/IF Activity filament Differentiated intermediate filament

Figure 4. Role of cadherin-based junctions and signaling in the formation of the tight junction (TJ)-containing granular layer. (A) Proposed model how asymmetric junction organization in SG2 promotes the formation of tension-high adherens junction, apically positioned tight junctions, and extended F-actin organization and adopt a shape resembling Kelvin’s tetracaidecahedron when SG3 cells become SG2 cells. Whereas in SG3 F-actin is attached through AJs at all cellular interfaces, in SG2 the apical F-actin network is uncoupled from the membrane due to absence of apical AJs. (B) Model integrating the role of adherens junctions (AJs), desmosomes and epidermal growth factor receptor (EGFR) in the regulation of actomyosin-dependent intercellular and cortical tension and TJ stability.

engage E-cadherin and Dsg1 of suprabasal cells. which we propose is essential to position cells Moreover, cytokinesis itself, through pulling suprabasally. forces of the cytokinetic ring, can locally alter and remodel neighboring junctions, promoting Transition of SG3 into a TJ Barrier self-organizing actin flows in these cells (Pin- Forming SG2 heiro et al. 2017) that likely further reinforce proper positioning of the two daughters either Similarly, the upward movement of individual basally or suprabasally (Fig. 3). Thus, both de- cells toward the granular layer (Rompolas et al. lamination and asymmetric division induce re- 2016) requires dynamic rearrangements of in- modeling of neighboring cell–cell interactions tercellular junctions. Whether this involves fast that spatiotemporally alter local cortical tension, cadherin turnover at the cell surface, local fast

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 15 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

exchange of adhesive and/or cytoskeletal con- of mechanical transition states in the epidermis. tacts, or other mechanisms is not known at But, do these transitions directly affect the dif- present. This transit through the different layers ferentiation status? Even though direct evidence also needs to be coupled to differentiation sta- is currently lacking, we propose that AJs and tus. As outlined above (see sections on “Polarity desmosomes synergistically coordinate epider- and Junctions” and “Epidermal Organization”), mal cell shape, position, and differentiation sta- recent results indicate the presence of a me- tus through integration of mechanical and chanical boundary between the SG3 and SG2 chemical signals such as tyrosine kinase receptor granular layer, as only these latter cells have signaling (see also Chiasson-MacKenzie and basolateral vinculin-positive AJs, apical barrier McClatchey 2017). As already discussed, Dsg1 forming TJs, and a strong increase in F-actin directly promotes differentiation by inhibiting organization (Rübsam et al. in press). These TJ- EGFR (Getsios et al. 2009), and several recent containing SG2 cells also adopt a defined shape observations suggest that this regulation likely that resembles a Kelvin’s tetracaidecahedron. involves AJs and cortical actomyosin activity Modelling showed that this specific shape en- (Erasmus et al. 2015; Broussard et al. 2017). ables TJ barrier maintenance when individual Firstly, E-cadherin has been implicated in regu- SG2 cells rearrange their contacts with their lating keratinocyte differentiation (Hines et al. SG2 neighbors to transit toward the SG1 (Yo- 1999; Charest et al. 2009) and, like loss of Dsg1, kouchi et al. 2016). The mechanism that con- epidermal loss of E-cadherin also promotes trols this mechanical transition and defined EGFR activity (Rübsam et al. in press). Interest- shape change are unclear but, as mentioned, ingly, differentiation is apparently normal in in part depend on E-cadherin and EGFR sig- these mice, perhaps because Dsg1 and P-cad- naling (Rübsam et al. in press). Of note, there is herin are up-regulated in these mice restricting a potential anisotropy of cortical tension in SG2 increased EGFR activity only to the basal layer cells. This is because SG2 cells only form AJs at (Tunggal et al. 2005). In agreement, removing all their basolateral but not at apical SG2–SG1 in- AJ mechanical signaling through the loss of α- terfaces, which contrasts with cells in the lower catenin promotes hyperproliferation and dis- layers, which uniformly connect all intercellular turbs tissue architecture associated with an in- interfaces to the keratin and F-actin cytoskele- crease in IGF-1R as well as Yap signaling (Va- ton through desmosomes and AJs, respectively. sioukhin et al. 2001a; Li et al. 2016). Moreover, We propose that the differential distribution of inactivation of myosin IIa in the epidermis not AJs and desmosomes in SG2 through synergis- only impairs TJs (Sumigray et al. 2012), but also tic regulation of intercellular and cortical ten- induces premature differentiation, suggesting sion, and tyrosine receptor kinase signaling cre- that the initial drop in tension observed when ate an asymmetry in junctional tension in SG2. cells initiate upward movement simultaneously This tension disequilibrium is necessary to pro- promotes delamination and differentiation (Le mote the formation of tension-high, vinculin- et al. 2016). Finally, cortical actomyosin organi- positive AJs and apical positioning of TJs the zation and activity regulates differentiation, TJs formation of tension-high AJs, apical TJs, and (Connelly et al. 2010; Zhou et al. 2013), and extended F-actin organization, which then col- EGFR activity status, whereas EGFR activity it- lectively drive the cell shape changes necessary self can determine tension states of cells and TJ to form tetracaidecahedron-shaped SG2 cells stability and function (Fig. 4B) (Rübsam et al. in (Fig. 4A). press). Together, these data suggest a model in which AJs and desmosomes cooperatively coordinate intercellular tension states with re- Integrating Junctional Control of Cell Shape, ceptor tyrosine kinase activity, which through Positioning, and Differentiation reinforcing feedback loops coordinate cell The above models implicate the synergistic ac- shape, position, barrier function, and differen- tion of cadherin-based junction as major drivers tiation status.

16 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

CONCLUDING REMARKS versity of Cologne, thus providing the basis for the concepts put forward in this review. The evolutionary invention of multicellular sheets undergoing morphogenetic movements required that cells mechanically and chemically REFERENCES interact. Classical cadherin/catenin adhesion ÃReference is also in this subject collection. complexes were instrumental in facilitating this development as these complexes integrated Abedin M, King N. 2008. The premetazoan ancestry of cad- through the formation of AJs three key proper- herins. Science 319: 946–948. ties: control of adhesion, cortical tension, and Abedin M, King N. 2010. Diverse evolutionary paths to cell adhesion. Trends Cell Biol 20: 734–742. signaling. The later arrival of desmosomes in Alegado RA, King N. 2014. Bacterial influences on animal vertebrates enabled organisms to not only me- origins. Cold Spring Harb Perspect Biol 6: a016162. chanically reinforce newly developed tissues Amack JD, Manning ML. 2012. Knowing the boundaries: such as the heart and skin epidermis, but also Extending the differential adhesion hypothesis in embry- onic cell sorting. Science 338: 212–215. provided novel regulatory opportunities to inte- Arimoto K, Burkart C, Yan M, Ran D, Weng S, Zhang DE. grate mechanical and chemical adhesion, cyto- 2014. Plakophilin-2 promotes tumor development by en- skeleton, and signaling networks necessary to hancing ligand-dependent and -independent epidermal accomplish increasing demands in terrestrial growth factor receptor dimerization and activation. Mol Cell Biol 34: 3843–3854. barrier function, immune regulation, and organ- Balda MS, Matter K. 2016. Tight junctions as regulators of ism/tissue size and complexity. Advances in tissue remodelling. Curr Opin Cell Biol 42: 94–101. high-resolution live imaging, optogenetics, and Bays JL, Campbell HK, Heidema C, Sebbagh M, DeMali KA. biophysical approaches will greatly facilitate fu- 2017. Linking E-cadherin mechanotransduction to cell metabolism through force-mediated activation of ture studies to determine in vivo junctional and AMPK. Nat Cell Biol 19: 724–731. tissue tension and monitor signaling and cell fate Blanpain C, Fuchs E. 2009. Epidermal homeostasis: A bal- in mammalian tissues to establish the principles ancing act of stem cells in the skin. Nat Rev Mol Cell Biol 10: – by which cadherin-based junctions coordinate 207 217. Boggetti B, Niessen CM. 2012. Adherens junctions in mam- spatiotemporal cell position, shape, and fate. malian development, homeostasis and disease: Lessons from mice. Subcell Biochem 60: 321–355. Bondow BJ, Faber ML, Wojta KJ, Walker EM, Battle MA. ACKNOWLEDGMENTS 2012. E-cadherin is required for intestinal morphogenesis in the mouse. Dev Biol 371: 1–12. We thank the Niessen, Green, and Wickström Bosher JM, Hahn BS, Legouis R, Sookhareea S, Weimer RM, laboratory members for many fruitful and in- Gansmuller A, Chisholm AD, Rose AM, Bessereau JL, Labouesse M. 2003. The Caenorhabditis elegans vab-10 spiring discussions as well as Alpha S. Yap for spectraplakin isoforms protect the epidermis against in- being supportive and providing great feedback. ternal and external forces. J Cell Biol 161: 757–768. The author’s work is supported by DFG SFB 829 Ã Braga V. 2017. Signaling by small GTPases at cell–cell junc- tions: Protein interactions building control and networks. A11, the Max Planck Society and Max Planck Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect. Foundation (to S.A.W.), National Institutes of a028746. Helath (NIH) Grants R01 AR041836, R37 Brennan D, Hu Y, Joubeh S, Choi YW, Whitaker-Menezes D, AR43380, R01 CA122151, and the J.L. Mayberry O’Brien T, Uitto J, Rodeck U, Mahoney MG. 2007. Supra- basal Dsg2 expression in transgenic mouse skin confers a Endowment (to K.J.G.) and German Research hyperproliferative and apoptosis-resistant phenotype to Foundation (DFG) Grants SFB 829 A1 and A5, keratinocytes. J Cell Sci 120: 758–771. SPP1782-NI1234/6–1 and the German Cancer Broussard JA, Getsios S, Green KJ. 2015. Desmosome regu- 360: – Aid (to C.M.N.). Dr. Green receives support as lation and signaling in disease. Cell Tissue Res 501 512. Associate Director of Basic Sciences in the Rob- Broussard JA, Yang R, Huang C, Nathamgari SSP, Beese AM, ert H Lurie Comprehensive Cancer Center of Godsel LM, Hegazy MH, Lee S, Zhou F, Sniadecki NJ, et Northwestern University (P30 CA60553). K.J. al. 2017. The desmoplakin/intermediate filament linkage regulates cell mechanics. Mol Biol Cell doi: 10.1091/mbc. G. and C.M.N. also thank the Alexander von E16-07-0520. Humboldt Foundation for supporting K.J.G. Buckley CD, Tan J, Anderson KL, Hanein D, Volkmann N, while she was performing research at the Uni- Weis WI, Nelson WJ, Dunn AR. 2014. Cell adhesion. The

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 17 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

minimal cadherin–catenin complex binds to actin fila- Erasmus JC, Welsh NJ, Braga VM. 2015. Cooperation of ments under force. Science 346: 1254211. distinct Rac-dependent pathways to stabilise E-cadherin Bulgakova NA, Brown NH. 2016. Drosophila p120–catenin adhesion. Cell Signal 27: 1905–1913. is crucial for endocytosis of the dynamic E-cadherin–Ba- Eshkind L, Tian Q, Schmidt A, Franke WW, Windoffer R, zooka complex. J Cell Sci 129: 477–482. Leube RE. 2002. Loss of desmoglein 2 suggests essential Candi E, Schmidt R, Melino G. 2005. The cornified envelope: functions for early embryonic development and prolifer- A model of cell death in the skin. Nat Rev Mol Cell Biol 6: ation of embryonal stem cells. Eur J Cell Biol 81: 592–598. 328–340. Fleming TP, Butler L, Lei X, Collins J, Javed Q, Sheth B, Carnahan RH, Rokas A, Gaucher EA, Reynolds AB. 2010. Stoddart N, Wild A, Hay M. 1994. Molecular maturation The molecular evolution of the p120–catenin subfamily of cell adhesion systems during mouse early development. and its functional associations. PLoS ONE 5: e15747. Histochemistry 101: 1–7. Charest JL, Jennings JM, King WP, Kowalczyk AP, Garcia AJ. Franke WW, Rickelt S, Barth M, Pieperhoff S. 2009. The 2009. Cadherin-mediated cell–cell contact regulates ker- junctions that don’t fit the scheme: Special symmetrical atinocyte differentiation. J Invest Dermatol 129: 564–572. cell-cell junctions of their own kind. Cell Tissue Res 338: – Ã Chiasson-MacKenzie C, McClatchey AI. 2017. Cell–cell 1 17. contact and receptor tyrosine kinase signaling. Cold Franzen CA, Todorovic V, Desai BV, Mirzoeva S, Yang XJ, Spring Harb Perspect Biol doi: 10.1101/cshperspect. Green KJ, Pelling JC. 2012. The desmosomal armadillo a029215. protein plakoglobin regulates prostate cancer cell adhe- sion and motility through vitronectin-dependent Src sig- Cirillo N, AlShwaimi E, McCullough M, Prime SS. 2014. 7: Pemphigus vulgaris autoimmune globulin induces Src- naling. PLoS ONE e42132. dependent tyrosine-phosphorylation of plakophilin 3 Fuchs E. 2007. Scratching the surface of skin development. and its detachment from desmoglein 3. Autoimmunity Nature 445: 834–842. 47: 134–140. Fujiwara M, Nagatomo A, Tsuda M, Obata S, Sakuma T, Connelly JT, Gautrot JE, Trappmann B, Tan DW, Donati G, Yamamoto T, Suzuki ST. 2015. Desmocollin-2 alone Huck WT, Watt FM. 2010. Actin and serum response forms functional desmosomal plaques, with the plaque factor transduce physical cues from the microenviron- formation requiring the juxtamembrane region and pla- ment to regulate epidermal stem cell fate decisions. Nat kophilins. J Biochem 158: 339–353. Cell Biol 12: 711–718. Furuse M, Hata M, Furuse K, Yoshida Y, Haratake A, Sugi- Conway DE, Coon BG, Budatha M, Arsenovic PT, Orsenigo tani Y, Noda T, Kubo A, Tsukita S. 2002. Claudin-based F, Wessel F, Zhang J, Zhuang Z, Dejana E, Vestweber D, et tight junctions are crucial for the mammalian epidermal al. 2017. VE-cadherin phosphorylation regulates endo- barrier: A lesson from claudin-1-deficient mice. J Cell Biol thelial fluid shear stress responses through the polarity 156: 1099–1111. protein LGN. Curr Biol 27: 2219–2225.e2215. Gallicano GI, Kouklis P, Bauer C, Yin M, Vasioukhin V, Cooper MD, Alder MN. 2006. The evolution of adaptive Degenstein L, Fuchs E. 1998. Desmoplakin is required immune systems. Cell 124: 815–822. early in development for assembly of desmosomes and 143: – Ã Delmar M, Laird DW, Naus CC, Nielsen MS, Versleis VK, cytoskeletal linkage. J Cell Biol 2009 2022. White TW. 2017. and disease. Cold Spring Ganeshan R, Chen J, Koch PJ. 2010. Mouse models for blis- Harb Perspect Biol doi: 10.1101/cshperspect.a029348. tering skin disorders. Dermatol Res Pract 2010: 584353. Delva E, Tucker DK, Kowalczyk AP. 2009. The desmosome. Garcia-Cattaneo A, Braga VM. 2013. Hold on tightly: How Cold Spring Harb Perspect Biol 1: a002543. to keep the local activation of small GTPases. Cell Adh 7: – Den Z, Cheng X, Merched-Sauvage M, Koch PJ. 2006. Des- Migr 283 287. mocollin 3 is required for pre-implantation development Garrod D, Tabernero L. 2014. Hyper-adhesion: A unique of the mouse embryo. J Cell Sci 119: 482–489. property of desmosomes. Cell Commun Adhes 21: 249– Dickinson DJ, Nelson WJ, Weis WI. 2011. A polarized ep- 256. ithelium organized by β- and α-catenin predates cadherin Getsios S, Simpson CL, Kojima S, Harmon R, Sheu LJ, Dusek and metazoan origins. Science 331: 1336–1339. RL, Cornwell M, Green KJ. 2009. Desmoglein 1-depen- Dickinson DJ, Nelson WJ, Weis WI. 2012a. An epithelial dent suppression of EGFR signaling promotes epidermal 185: – tissue in Dictyostelium challenges the traditional origin differentiation and morphogenesis. J Cell Biol 1243 of metazoan multicellularity. Bioessays 34: 833–840. 1258. Dickinson DJ, Robinson DN, Nelson WJ, Weis WI. 2012b. Godsel LM, Dubash AD, Bass-Zubek AE, Amargo EV, α-Catenin and IQGAP regulate myosin localization to Klessner JL, Hobbs RP, Chen X, Green KJ. 2010. Plako- control epithelial tube morphogenesis in Dictyostelium. philin 2 couples actomyosin remodeling to desmosomal 21: – Dev Cell 23: 533–546. plaque assembly via RhoA. Mol Biol Cell 2844 2859. Dubash AD, Koetsier JL, Amargo EV, Najor NA, Harmon Goonesinghe A, Luan XM, Hurlstone A, Garrod D. 2012. fi RM, Green KJ. 2013. The GEF Bcr activates RhoA/MAL Desmosomal cadherins in zebra sh epiboly and gastru- 12: signaling to promote keratinocyte differentiation via des- lation. BMC Dev Biol 1. moglein-1. J Cell Biol 202: 653–666. Green KJ, Getsios S, Troyanovsky S, Godsel LM. 2010. In- Elias PM, Matsuyoshi N, Wu H, Lin C, Wang ZH, Brown BE, tercellular junction assembly, dynamics, and homeosta- 2: Stanley JR. 2001. Desmoglein isoform distribution affects sis. Cold Spring Harb Perspect Biol a000125. stratum corneum structure and function. J Cell Biol 153: Grimson MJ, Coates JC, Reynolds JP, Shipman M, Blanton 243–249. RL, Harwood AJ. 2000. Adherens junctions and β-cate-

18 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

nin-mediated cell signalling in a non-metazoan organ- King N. 2004. The unicellular ancestry of animal develop- ism. Nature 408: 727–731. ment. Dev Cell 7: 313–325. Gul IS, Hulpiau P, Saeys Y, van Roy F. 2017. Metazoan evo- Klessner JL, Desai BV, Amargo EV, Getsios S, Green KJ. lution of the armadillo repeat superfamily. Cell Mol Life 2009. EGFR and ADAMs cooperate to regulate shedding Sci 74: 525–541. and endocytic trafficking of the desmosomal cadherin Gumbiner B, Stevenson B, Grimaldi A. 1988. The role of the desmoglein 2. Mol Biol Cell 20: 328–337. uvomorulin in the formation and Koetsier JL, Amargo EV, Todorovic V, Green KJ, Godsel LM. maintenance of the epithelial junctional complex. J Cell 2014. Plakophilin 2 affects cell migration by modulating Biol 107: 1575–1587. dynamics and integrin protein expression. Guo Z, Neilson LJ, Zhong H, Murray PS, Zanivan S, Zaidel- J Invest Dermatol 134: 112–122. Bar R. 2014. E-cadherin interactome complexity and ro- Kourtidis A, Ngok SP, Anastasiadis PZ. 2013. p120 catenin: bustness resolved by quantitative proteomics. Sci Signal 7: An essential regulator of cadherin stability, adhesion-in- rs7. duced signaling, and cancer progression. Prog Mol Biol Hardin J. 2015. Getting to the core of cadherin complex Transl Sci 116: 409–432. function in Caenorhabditis elegans. F1000Res doi: Kubo A, Nagao K, Amagai M. 2012. Epidermal barrier dys- 10.12688/f1000research.6866.1. function and cutaneous sensitization in atopic diseases. J Harmon RM, Simpson CL, Johnson JL, Koetsier JL, Dubash Clin Invest 122: 440–447. AD, Najor NA, Sarig O, Sprecher E, Green KJ. 2013. Ladoux B, Nelson WJ, Yan J, Mege RM. 2015. The mecha- Desmoglein-1/erbin interaction suppresses ERK activa- notransduction machinery at work at adherens junctions. tion to support epidermal differentiation. J Clin Invest Integr Biol (Camb) 7: 1109–1119. 123: – 1556 1570. Lai-Cheong JE, Arita K, McGrath JA. 2007. Genetic diseases Harris TJ, Tepass U. 2010. Adherens junctions: From mol- of junctions. J Invest Dermatol 127: 2713–2725. 11: – ecules to morphogenesis. Nat Rev Mol Cell Biol 502 Le HQ, Ghatak S, Yeung CC, Tellkamp F, Gunschmann C, 514. Dieterich C, Yeroslaviz A, Habermann B, Pombo A, Nies- Hart KC, Tan J, Siemers KA, Sim JY, Pruitt BL, Nelson WJ, sen CM, et al. 2016. Mechanical regulation of transcrip- Gloerich M. 2017. E-cadherin and LGN align epithelial tion controls Polycomb-mediated gene silencing during cell divisions with tissue tension independently of cell lineage commitment. Nat Cell Biol 18: 864–875. 114: – shape. Proc Natl Acad Sci E5845 E5853. Leckband DE, de Rooij J. 2014. Cadherin adhesion and me- Hatzfeld M, Wolf A, Keil R. 2014. Plakophilins in desmoso- chanotransduction. Annu Rev Cell Dev Biol 30: 291–315. 21: – mal adhesion and signaling. Cell Commun Adhes 25 Lenox JM, Koch PJ, Mahoney MG, Lieberman M, Stanley JR, 42. Radice GL. 2000. Postnatal lethality of P-cadherin/des- Ã Hatzfeld M, Keil R, Magin TM. 2017. Desmosomes and moglein 3 double knockout mice: Demonstration of a intermediate filaments: Their consequences for tissue me- cooperative effect of these cell adhesion molecules in tis- chanics. Cold Spring Harb Perspect Biol doi: 10.1101/ sue homeostasis of stratified squamous epithelia. J Invest cshperspect.a029157. Dermatol 114: 948–952. Heisenberg CP, Bellaiche Y. 2013. Forces in tissue morpho- Leung CL, Green KJ, Liem RK. 2002. Plakins: A family of genesis and patterning. Cell 153: 948–962. versatile cytolinker proteins. Trends Cell Biol 12: 37–45. Henkler F, Strom M, Mathers K, Cordingley H, Sullivan K, Li P, Silvis MR, Honaker Y, Lien WH, Arron ST, Vasioukhin King I. 2001. Trangenic misexpression of the differentia- V. 2016. αE-catenin inhibits a Src–YAP1 oncogenic mod- tion-specific desmocollin isoform 1 in basal keratino- ule that couples tyrosine kinases and the effector of Hippo cytes. J Invest Dermatol 116: 144–149. signaling pathway. Genes Dev 30: 798–811. Hines MD, Jin HC, Wheelock MJ, Jensen PJ. 1999. Inhibi- Lin S, Gordon K, Kaplan N, Getsios S. 2010. Ligand targeting tion of cadherin function differentially affects markers of of EphA2 enhances keratinocyte adhesion and differen- terminal differentiation in cultured human keratinocytes. tiation via desmoglein 1. Mol Biol Cell 21: 3902–3914. 112: – J Cell Sci 4569 4579. Lyon RC, Mezzano V, Wright AT, Pfeiffer E, Chuang J, Huelsmann S, Brown NH. 2014. Spectraplakins. Curr Biol Banares K, Castaneda A, Ouyang K, Cui L, Contu R, et 24: R307–R308. al. 2014. Connexin defects underlie arrhythmogenic right Hulpiau P, Gul IS, van Roy F. 2013. New insights into the ventricular cardiomyopathy in a novel mouse model. evolution of metazoan cadherins and catenins. Prog Mol Hum Mol Genet 23: 1134–1150. Biol Transl Sci 116: 71–94. McAleer MA, Pohler E, Smith FJ, Wilson NJ, Cole C, Mac- Jamora C, DasGupta R, Kocieniewski P, Fuchs E. 2003. Links Gowan S, Koetsier JL, Godsel LM, Harmon RM, Gruber between signal transduction, transcription and adhesion R, et al. 2015. Severe dermatitis, multiple allergies, and in epithelial bud development. Nature 422: 317–322. metabolic wasting syndrome caused by a novel mutation Kamekura R, Kolegraff KN, Nava P, Hilgarth RS, Feng M, in the N-terminal plakin domain of desmoplakin. J Aller- 136: – Parkos CA, Nusrat A. 2014. Loss of the desmosomal cad- gy Clin Immunol 1268 1276. herin desmoglein-2 suppresses colon cancer cell prolifer- McCauley MD, Wehrens XH. 2009. Animal models of ar- ation through EGFR signaling. Oncogene 33: 4531–4536. rhythmogenic cardiomyopathy. Dis Model Mech 2: 563– Kant S, Holthofer B, Magin TM, Krusche CA, Leube RE. 570. 2015. Desmoglein 2-dependent arrhythmogenic cardio- McCrea PD, Gottardi CJ. 2016. Beyond β-catenin: Prospects myopathy is caused by a loss of adhesive function. Circ for a larger catenin network in the nucleus. Nat Rev Mol Cardiovasc Genet 8: 553–563. Cell Biol 17: 55–64.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 19 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

M. Rübsam et al.

à Mege RM, Ishiyama N. 2017. Integration of cadherin adhe- Pizarro-Cerda J, Kuhbacher A, Cossart P. 2012. Entry of sion and cytoskeleton at adherens junctions. Cold Spring Listeria monocytogenes in mammalian epithelial cells: Harb Perspect Biol doi: 10.1101/cshperspect.a028738. An updated view. Cold Spring Harb Perspect Med 2: Merritt AJ, Berika MY, Zhai W, Kirk SE, Ji B, Hardman MJ, a010009. Garrod DR. 2002. Suprabasal desmoglein 3 expression in Posern G, Treisman R. 2006. Actin’ together: Serum re- the epidermis of transgenic mice results in hyperprolifer- sponse factor, its cofactors and the link to signal trans- ation and abnormal differentiation. Mol Cell Biol 22: duction. Trends Cell Biol 16: 588–596. 5846–5858. Ramms L, Fabris G, Windoffer R, Schwarz N, Springer R, Michels C, Buchta T, Bloch W, Krieg T, Niessen CM. 2009. Zhou C, Lazar J, Stiefel S, Hersch N, Schnakenberg U, et Classical cadherins regulate desmosome formation. J In- al. 2013. Keratins as the main component for the mechan- vest Dermatol 129: 2072–2075. ical integrity of keratinocytes. Proc Natl Acad Sci 110: Miller PW, Clarke DN, Weis WI, Lowe CJ, Nelson WJ. 2013. 18513–18518. The evolutionary origin of epithelial cell–cell adhesion Ray S, Foote HP, Lechler T. 2013. β-Catenin protects the mechanisms. Curr Top Membr 72: 267–311. epidermis from mechanical stresses. J Cell Biol 202: 45– Muroyama A, Lechler T. 2012. Polarity and stratification of 52. the epidermis. Semin Cell Dev Biol 23: 890–896. Rodriguez-Boulan E, Macara IG. 2014. Organization and Murray PS, Zaidel-Bar R. 2014. Pre-metazoan origins and execution of the programme. Nat Rev evolution of the cadherin adhesome. Biol Open 3: 1183– Mol Cell Biol 15: 225–242. 1195. Rompolas P, Mesa KR, Kawaguchi K, Park S, Gonzalez D, Myster SH, Cavallo R, Anderson CT, Fox DT, Peifer M. 2003. Brown S, Boucher J, Klein AM, Greco V. 2016. Spatio- Drosophila p120catenin plays a supporting role in cell temporal coordination of stem cell commitment during adhesion but is not an essential adherens junction com- epidermal homeostasis. Science 352: 1471–1474. ponent. J Cell Biol 160: 433–449. Rübsam M, Mertz AF, Kubo A, Marg S, Jüngst C, Goranci- Newman SA. 2016. ‘Biogeneric’ developmental processes: Buzhala G, Schauss A, Horsley V, Dufresne ER, Moser M, Drivers of major transitions in animal evolution. Philos et al. E-cadherin integrates mechanotransduction and Trans R Soc Lond B Biol Sci 371: 20150443. EGFR signaling to control junctional tissue polarization Nichols SA, Roberts BW, Richter DJ, Fairclough SR, King N. and Tight Junction positioning. Nat Commun. (In press). 2012. Origin of metazoan cadherin diversity and the an- Samuelov L, Sprecher E. 2015. Inherited desmosomal disor- tiquity of the classical cadherin/β–catenin complex. Proc ders. Cell Tissue Res 360: 457–475. Natl Acad Sci 109: 13046–13051. Samuelov L, Sarig O, Harmon RM, Rapaport D, Ishida-Ya- Nielsen MS, Axelsen LN, Sorgen PL, Verma V, Delmar M, mamoto A, Isakov O, Koetsier JL, Gat A, Goldberg I, Holstein-Rathlou NH. 2012. Gap junctions. Compr Phys- Bergman R, et al. 2013. Desmoglein 1 deficiency results iol 2: 1981–2035. in severe dermatitis, multiple allergies and metabolic 45: – Niessen CM, Leckband D, Yap AS. 2011. Tissue organization wasting. Nat Genet 1244 1248. by cadherin adhesion molecules: Dynamic molecular and Schlegelmilch K, Mohseni M, Kirak O, Pruszak J, Rodriguez cellular mechanisms of morphogenetic regulation. Phys- JR, Zhou D, Kreger BT, Vasioukhin V, Avruch J, Brum- iol Rev 91: 691–731. melkamp TR, et al. 2011. Yap1 acts downstream of α- Oda H, Takeichi M. 2011. Evolution: Structural and func- catenin to control epidermal proliferation. Cell 144: tional diversity of cadherin at the adherens junction. J Cell 782–795. Biol 193: 1137–1146. Schneider MR, Dahlhoff M, Horst D, Hirschi B, Trulzsch K, Olson EN. 2006. Gene regulatory networks in the evolution Muller-Hocker J, Vogelmann R, Allgauer M, Gerhard M, and development of the heart. Science 313: 1922–1927. Steininger S, et al. 2010. A key role for E-cadherin in δ intestinal homeostasis and Paneth cell maturation. PLoS Pacquelet A, Lin L, Rorth P. 2003. Binding site for p120/ - 5: catenin is not required for Drosophila E-cadherin func- ONE e14325. tion in vivo. J Cell Biol 160: 313–319. Seltmann K, Fritsch AW, Kas JA, Magin TM. 2013. Keratins fi Padmanabhan A, Rao MV, Wu Y, Zaidel-Bar R. 2015. Jack of signi cantly contribute to cell stiffness and impact inva- 110: – all trades: Functional modularity in the adherens junc- sive behavior. Proc Natl Acad Sci 18507 18512. tion. Curr Opin Cell Biol 36: 32–40. Simpson CL, Patel DM, Green KJ. 2011. Deconstructing the Patel DM, Dubash AD, Kreitzer G, Green KJ. 2014. Disease skin: Cytoarchitectural determinants of epidermal mor- 12: – mutations in desmoplakin inhibit Cx43 membrane tar- phogenesis. Nat Rev Mol Cell Biol 565 580. geting mediated by desmoplakin–EB1 interactions. J Cell Siu CH, Harris TJ, Wang J, Wong E. 2004. Regulation of cell– Biol 206: 779–797. cell adhesion during Dictyostelium development. Semin 15: – Pinheiro D, Hannezo E, Herszterg S, Bosveld F, Gaugue I, Cell Dev Biol 633 641. Balakireva M, Wang Z, Cristo I, Rigaud SU, Markova O, et Sumigray KD, Lechler T. 2012. Desmoplakin controls mi- al. 2017. Transmission of cytokinesis forces via E-cad- crovilli length but not cell adhesion or keratin organiza- herin dilution and actomyosin flows. Nature 545: 103– tion in the . Mol Biol Cell 23: 792– 107. 799. Pires-daSilva A, Sommer RJ. 2003. The evolution of signal- Sumigray KD, Chen H, Lechler T. 2011. Lis1 is essential for ling pathways in animal development. Nat Rev Genet 4: cortical microtubule organization and desmosome stabil- 39–49. ity in the epidermis. J Cell Biol 194: 631–642.

20 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Evolutionary Perspective of Adherens Junctions and Desmosomes

Sumigray KD, Foote HP, Lechler T. 2012. Noncentrosomal Vasioukhin V, Bowers E, Bauer C, Degenstein L, Fuchs E. microtubules and type II myosins potentiate epidermal 2001b. Desmoplakin is essential in epidermal sheet for- cell adhesion and barrier formation. J Cell Biol 199: 513– mation. Nat Cell Biol 3: 1076–1085. 525. Vite A, Radice GL. 2014. N-cadherin/catenin complex as a – Sumigray K, Zhou K, Lechler T. 2014. Cell cell adhesions master regulator of function. Cell Com- and cell contractility are upregulated upon desmosome mun Adhes 21: 169–179. disruption. PLoS ONE 9: e101824. Walko G, Woodhouse S, Pisco AO, Rognoni E, Liakath-Ali Tellkamp F, Vorhagen S, Niessen CM. 2014. Epidermal po- K, Lichtenberger BM, Mishra A, Telerman SB, Viswana- larity genes in health and disease. Cold Spring Harb Per- than P, Logtenberg M, et al. 2017. A genome-wide screen spect Med 4: a015255. identifies YAP/WBP2 interplay conferring growth advan- Thomason HA, Scothern A, McHarg S, Garrod DR. 2010. tage on human epidermal stem cells. Nat Commun 8: Desmosomes: Adhesive strength and signalling in health 14744. and disease. Biochem J 429: 419–433. Watt FM. 2014. Mammalian skin cell biology: At the inter- Tinkle CL, Pasolli HA, Stokes N, Fuchs E. 2008. New insights face between laboratory and clinic. Science 346: 937–940. into cadherin function in epidermal sheet formation and maintenance of tissue integrity. Proc Natl Acad Sci 105: Ã Yap AS, Duszyc K, Viasnoff V. 2017. Mechanosensing and 15405–15410. mechanotransduction at cell–cell junctions. Cold Spring Todorovic V, Desai BV, Patterson MJ, Amargo EV, Dubash Harb Perspect Biol doi: 10.1101/cshperspect.a028761. AD, Yin T, Jones JC, Green KJ. 2010. Plakoglobin regu- Yokouchi M, Atsugi T, Logtestijn MV, Tanaka RJ, Kajimura lates cell motility through Rho- and fibronectin-depen- M, Suematsu M, Furuse M, Amagai M, Kubo A. 2016. dent Src signaling. J Cell Sci 123: 3576–3586. Epidermal cell turnover across tight junctions based on Tunggal JA, Helfrich I, Schmitz A, Schwarz H, Gunzel D, Kelvin’s tetrakaidecahedron cell shape. Elife 5: e19593. Fromm M, Kemler R, Krieg T, Niessen CM. 2005. E-cad- Zhang H, Pasolli HA, Fuchs E. 2011. Yes-associated protein herin is essential for in vivo epidermal barrier function by (YAP) transcriptional coactivator functions in balancing 24: – regulating tight junctions. EMBO J 1146 1156. growth and differentiation in skin. Proc Natl Acad Sci 108: Turksen K, Troy TC. 2002. Permeability barrier dysfunction 2270–2275. in transgenic mice overexpressing claudin 6. Development Zhao ZM, Reynolds AB, Gaucher EA. 2011. The evolution- 129: 1775–1784. ary history of the catenin gene family during metazoan Van Itallie CM, Anderson JM. 2014. Architecture of tight evolution. BMC Evol Biol 11: 198. junctions and principles of molecular composition. Zhou K, Muroyama A, Underwood J, Leylek R, Ray S, So- Semin Cell Dev Biol 36: 157–165. derling SH, Lechler T. 2013. Actin-related protein2/3 Vasioukhin V, Bauer C, Degenstein L, Wise B, Fuchs E. complex regulates tight junctions and terminal differen- 2001a. Hyperproliferation and defects in epithelial polar- α tiation to promote epidermal barrier formation. Proc Natl ity upon conditional ablation of -catenin in skin. Cell 110: – 104: 605–617. Acad Sci E3820 E3829.

Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029207 21 Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Adherens Junctions and Desmosomes Coordinate Mechanics and Signaling to Orchestrate Tissue Morphogenesis and Function: An Evolutionary Perspective

Matthias Rübsam, Joshua A. Broussard, Sara A. Wickström, Oxana Nekrasova, Kathleen J. Green and Carien M. Niessen

Cold Spring Harb Perspect Biol published online September 11, 2017

Subject Collection Cell-Cell Junctions

Vascular Endothelial (VE)-Cadherin, Endothelial Signaling by Small GTPases at Cell−Cell Adherens Junctions, and Vascular Disease Junctions: Protein Interactions Building Control Maria Grazia Lampugnani, Elisabetta Dejana and and Networks Costanza Giampietro Vania Braga Adherens Junctions and Desmosomes Making Connections: Guidance Cues and Coordinate Mechanics and Signaling to Receptors at Nonneural Cell−Cell Junctions Orchestrate Tissue Morphogenesis and Function: Ian V. Beamish, Lindsay Hinck and Timothy E. An Evolutionary Perspective Kennedy Matthias Rübsam, Joshua A. Broussard, Sara A. Wickström, et al. Cell−Cell Contact and Receptor Tyrosine Kinase The Cadherin Superfamily in Neural Circuit Signaling Assembly Christine Chiasson-MacKenzie and Andrea I. James D. Jontes McClatchey Hold Me, but Not Too Tight−−Endothelial Cell−Cell Mechanosensing and Mechanotransduction at Junctions in Angiogenesis Cell−Cell Junctions Anna Szymborska and Holger Gerhardt Alpha S. Yap, Kinga Duszyc and Virgile Viasnoff Connexins and Disease Beyond Cell−Cell Adhesion: Sensational Mario Delmar, Dale W. Laird, Christian C. Naus, et Cadherins for Hearing and Balance al. Avinash Jaiganesh, Yoshie Narui, Raul Araya-Secchi, et al. Cell Junctions in Hippo Signaling Cell−Cell Junctions Organize Structural and Ruchan Karaman and Georg Halder Signaling Networks Miguel A. Garcia, W. James Nelson and Natalie Chavez

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved Downloaded from http://cshperspectives.cshlp.org/ on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press

Loss of E-Cadherin-Dependent Cell−Cell Adhesion Cell Biology of Tight Junction Barrier Regulation and the Development and Progression of Cancer and Mucosal Disease Heather C. Bruner and Patrick W.B. Derksen Aaron Buckley and Jerrold R. Turner Desmosomes and Intermediate Filaments: Their Integration of Cadherin Adhesion and Consequences for Tissue Mechanics Cytoskeleton at Adherens Junctions Mechthild Hatzfeld, René Keil and Thomas M. René Marc Mège and Noboru Ishiyama Magin

For additional articles in this collection, see http://cshperspectives.cshlp.org/cgi/collection/

Copyright © 2017 Cold Spring Harbor Laboratory Press; all rights reserved