Epithelial cells form tissues that generate biological barriers in the body. Tight junctions (TJs) are responsible for maintaining a selectively permeable seal between epithelial cells, but little is known about how TJs dynamically remodel in response to physiological forces that challenge epithelial barrier function, such as cell shape changes (e.g. during cell division) or tissue stretching (e.g. during developmental morphogenesis). In this Review, we first introduce a framework to think about TJ remodeling across multiple scales: from molecular dynamics, to strand dynamics, to cell- and tissue-scale dynamics. We then relate knowledge gained from global perturbations of TJs to emerging information about local TJ remodeling events, where transient localized Rho activation and actomyosin-mediated contraction promote TJ remodeling to repair local leaks in barrier function. We conclude by identifying emerging areas in the field and propose ideas for future studies that address unanswered questions about the mechanisms that drive TJ remodeling.

Epithelia aid in organizing the body by creating tissue barriers – sheets of connected cells with the ability to regulate the quantity and type of materials that cross the tissue. In addition to acting as barriers, epithelial tissues are involved in specialized functions, such as absorption, secretion, transportation and signaling. Almost every organ in our body is covered with or lined by an epithelial tissue that is specifically suited for the task of the organ.

Molecules can cross tissues by going either through cells (transcellular transport) or between cells (paracellular transport), and different tissues have different requirements for the strength and selectivity of these barriers. Tight junctions (TJs) are responsible for sealing the paracellular space and discriminating among small solutes, such as ions, water and small uncharged molecules, while generally restricting the passage of larger molecules and microorganisms (Marchiando et al., 2010). These barriers are crucial for establishing concentration gradients across tissues, thereby creating specialized environments (e.g. a low pH in the stomach), and preventing infection.

TJs regulate the flux of molecules through the paracellular spaces with a family of transmembrane proteins called claudins, which are capable of forming size- and charge-selective pores, as well as restricting flux. Vertebrate genomes encode a large number of claudins (more than 20 in humans) with diverse pore- and barrier-forming properties (Günzel and Yu, 2013 ; see also Box 1). TJs are connected to the actomyosin cytoskeleton through scaffolding proteins, including ZO family proteins (Box 1), and these cytoskeletal connections are crucial for the structure and function of TJs. The organization and tension of perijunctional actomyosin is regulated by the small GTPase RhoA; thus, RhoA activity must be tightly regulated in space and time for correct TJ formation, maintenance and remodeling (Arnold et al., 2017).

Box 1. Molecular organization of TJs

The claudin family of transmembrane proteins (yellow and orange), comprising >20 members, oligomerize into strands and interlock with their counterparts on neighboring cells to selectively regulate passage through the paracellular space. Whereas ‘barrier-forming’ claudins primarily restrict ion flux (i.e. claudins 1, 3, 5, 6, 9, 14 and 19), ‘pore-forming’ claudins facilitate it by forming size- and charge-specific pores (i.e. claudins 2, 10a, 10b, 15, 17 and 21) (Gunzel and Yu, 2013). Together with claudin strands, junctional adhesion molecules (JAMs, dark blue) also help limit macromolecule flux (Otani et al., 2019). TJ-associated MARVEL proteins (TAMPs, green), such as occludin, tricellulin and MarvelD3, play a variety of roles in signaling, barrier development and TJ strand morphology (Raleigh et al., 2010; Cording et al., 2013; Krug et al., 2009).

ZO proteins (light blue) facilitate the formation of a dense plaque of proteins associated with TJs (Beutel et al., 2019; Otani et al., 2019). A ZO protein can multimerize with itself and other ZO proteins (e.g. ZO-1 can bind ZO-2 and ZO-3 in addition to itself), and can bind claudins, JAMs, TAMPs, F-actin (red), scaffolding proteins (e.g. cingulin, pink) and signaling molecules (not illustrated), including GEFs (e.g. Tuba) and transcription factors (e.g. ZONAB) (Shen et al., 2011). For a detailed summary of protein-protein interactions within the TJ, see Shen et al. (2011).

Pore and leak pathways for paracellular permeability

TJ selectivity is regulated through claudin pores – channels formed by the extracellular domains of claudins – that conduct ions and small molecules on the basis of size and charge. Paracellular permeability through claudin pores is commonly referred to as the ‘pore pathway’. Selectivity is based on the composition of pore-forming claudins (both cation and anion pores) and barrier-forming claudins, which restrict the flux of small solutes (Günzel, 2017). Barrier properties of TJs can change in response to various stimuli by ‘claudin switching’, e.g. by replacing a pore-forming claudin with a barrier-forming claudin (Günzel, 2017). Owing to the large number of highly selective pores, the pore pathway is characterized by its high capacity and high selectivity (Anderson and Van Itallie, 2009; Shen et al., 2011). Transepithelial electrical resistance (TER) measures the magnitude of ion flux across epithelia (increased TER indicates decreased ion permeability), whereas more-refined techniques can measure size and charge selectivity of junctions (Box 2; see also Shen et al., 2011).

Box 2. Pore and leak pathways of paracellular permeability

Depending on their permeability characteristics, TJ barriers can broadly be divided into two categories: (i) the ‘pore pathway’, with permeability for ions and small solutes with a radius of ≤4Å (Van Itallie et al., 2008) and, (ii) the ‘leak pathway’, with permeability for larger (≥4Å) solutes and macromolecules. The barrier to ions is often measured using transepithelial electrical resistance (TER), where electrical current is generated across a monolayer, the voltage is measured and the resistance calculated (Srinivasan et al., 2015). Because cell membranes are good resistors, the electrical current will be carried by ion flux through the paracellular route (Srinivasan et al., 2015). In intact TJs, ion flux will occur through claudin pores. Thus, tissues with a high proportion of barrier-forming claudins, such as bladder and stomach, have higher electrical resistance (see Figure, top left and bottom left), whereas tissues with a high proportion of pore-forming claudins, such as kidney proximal tubule and small intestine, exhibit low electrical resistance (see Figure top right and bottom right). In some situations, tissues can exhibit high macromolecular flux, yet, also high electrical resistance (see Figure, bottom left) (Balda et al., 1996). This paradoxical finding helped researchers to recognize that there must be a second pathway of paracellular permeability: the leak pathway. Notably, high ion flux can also occur at sites where the strand network is disrupted (see Figure, bottom right). Thus, it is important to note that a drop in TER can indicate either an increase in ion flux owing to TJ disruption or an increase in the incorporation and/or opening of pore-forming claudins. To aid in the interpretation of a drop in TER, changes in TJ size and charge selectivity could be measured with methods such as bi-ionic substitution or dilution potential (e.g. Raleigh et al., 2011).

Macromolecular flux is typically measured after the addition of traceable macromolecules, such as fluorescent dextrans or PEG oligomers, to one side of the epithelium and following their passage to the other side. PEG oligomers are available in sizes small enough to allow measurement of the upper radius of molecules that travel through the pore pathway, as well as those that use the leak pathway (Van Itallie et al., 2008; Watson et al., 2001). Fluorescent dextrans at different sizes can also be used to measure the size selectivity of the leak pathway (e.g. Otani et al., 2019). However, there are limitations with these techniques, which include averaging of the barrier properties across the entire tissue, preventing one to understand how leaks are affected by changes in cell morphology or cell position in the epithelium. Whereas fluorescent dextrans can be visualized in vivo (e.g. Higashi et al., 2016), the sensitivity of this method might not be sufficient to detect small volumes of molecules breaching the epithelium. More-sensitive assays, such as the highly sensitive Zinc-based ultrasensitive microscopic barrier assay (ZnUMBA) and avidin–biotin sandwich assays (Stephenson et al., 2019; Richter et al., 2016), allow for finer spatial and temporal resolution of flux through the leak pathway.

A second pathway, known as the ‘leak pathway’, allows larger molecules to cross TJs, albeit with less selectivity and a much lower capacity than the pore pathway (Shen et al., 2011). The predominate hypothesis is that leak pathway flux occurs due to the breaking and annealing of claudin strands, which may be influenced by intermolecular associations between claudins, occludin, ZO-1 and the actin cytoskeleton (Fanning et al., 2012; Turner et al., 2014; Van Itallie et al., 2015). Another leak pathway regulator, JAM-A, is sufficient to limit the flux of macromolecules in the absence of claudin strands, suggesting that JAMs serve as a stop-gap measure to protect barrier integrity when claudin strands are disrupted or newly forming (Otani et al., 2019). The presence of redundant mechanisms to regulate the leak pathway underlines the importance of restricting flux of macromolecules via the leak pathway.

Cell shape changes challenge epithelial barrier function

To effectively limit flux, it is necessary for TJs to be continuous around the perimeter of epithelial cells. However, multiple physiological processes in epithelial tissues involve cell-shape changes that require cell–cell boundaries to expand and contract, thus posing challenges to the continuity of TJs. For example, actomyosin-driven changes of cell shape and cell rearrangements induce the stretching, bending and folding of tissues during developmental morphogenesis (Harris and Tepass, 2010). Other mechanical challenges arise from organ-specific function, including bladder expansion and gut peristalsis. However, it is poorly understood how such dynamic tissues can also act as stable barriers. Because the correct regulation of barrier function is essential for optimal organ function (Marchiando et al., 2010), epithelial cells must possess robust mechanisms for maintaining barrier function while remaining plastic enough to adapt to a changing environment. This Review focuses on how TJ remodeling on several scales contributes to sustained barrier function in the face of cell and tissue dynamics.

TJ dynamics encompass multiple scales – from the molecular composition and dynamics of individual proteins and protein complexes to tissue-scale events, such as mechanical stress – which affect TJ remodeling (Fig. 1). Molecular- and tissue-scale dynamics of TJs have been studied in some detail, although our knowledge of these processes is still incomplete. An even greater gap in knowledge lies in the intermediate scales – at the strand network and cellular scales (Fig. 1). In this section, we will discuss TJ dynamics across scales, the technical limitations that prevent us from fully understanding these remarkable structures, and the interplay between these levels of TJ organization and remodeling.

Fig. 1.

TJs are dynamic at the molecular, strand, cellular and tissue scale. Molecular dynamics include the trafficking of proteins to the TJ, the formation of protein complexes and incorporation of claudins into strands. Strand dynamics include the assembly of strands into networks, dynamic reorganization of these networks at baseline and in response to stimuli, change in network geometry, and the incorporation and removal of claudins from strands. At the cellular scale, TJs undergo local alterations as cell–cell boundaries elongate, shrink or otherwise experience a change in shape, such as during cell division, extrusion or axis elongation. At the tissue scale, TJs change in response to tissue-scale forces (e.g. mechanical forces associated with development or organ function), biochemical signals (e.g. immune response), and differentiation. In turn, the state of the tissue influences the expression and trafficking of different TJ components, ultimately altering the molecular, strand and cellular dynamics of TJs.

Fig. 1.

TJs are dynamic at the molecular, strand, cellular and tissue scale. Molecular dynamics include the trafficking of proteins to the TJ, the formation of protein complexes and incorporation of claudins into strands. Strand dynamics include the assembly of strands into networks, dynamic reorganization of these networks at baseline and in response to stimuli, change in network geometry, and the incorporation and removal of claudins from strands. At the cellular scale, TJs undergo local alterations as cell–cell boundaries elongate, shrink or otherwise experience a change in shape, such as during cell division, extrusion or axis elongation. At the tissue scale, TJs change in response to tissue-scale forces (e.g. mechanical forces associated with development or organ function), biochemical signals (e.g. immune response), and differentiation. In turn, the state of the tissue influences the expression and trafficking of different TJ components, ultimately altering the molecular, strand and cellular dynamics of TJs.

Strand dynamics

The basic unit of the TJ is the claudin-based strand (Fig. 1, strands). Multiple claudin-based strands join together to form elaborate strand networks that vary considerably in complexity and organization from tissue to tissue (Claude and Goodenough, 1973). Because claudins possess diverse pore-forming and barrier-forming properties (Günzel and Yu, 2013; see also Box 1), the composition of claudins within the strand network has a profound effect on ion flux across the tissue (Fig. 1, molecules and strands; Box 2). Less well-understood is how the morphology of the strand network itself impacts barrier function.

Owing to the tight packing of TJ strands and the relatively poor z-resolution of light microscopy, the morphology of the TJ strand network has traditionally been viewed by freeze fracture electron microscopy (FFEM). One FFEM study comparing the epithelial strand networks that exhibit different barrier properties found that an increased strand number is generally associated with decreased ion flux (Claude and Goodenough, 1973). However, there are also examples where TJ permeability is independent of strand number (Colegio et al., 2003), suggesting the claudin composition – not just the number of strands – determines permeability.

Some treatments that affect barrier properties correspond to rapid changes in strand network configuration. For example, exposure of Necturus (salamander) gallbladder to plant cytokinins causes a rapid but reversible increase in TER (Bentzel et al., 1976; see also Box 2). This treatment also results in the reorganization of TJ strand networks from being mostly parallel relative to the apical membrane to more-disorganized, vertically oriented networks with a higher number of free strand ends (Bentzel et al., 1980). Removal of cytokinins corresponds to a drop in TER and a reorientation of strands to a parallel orientation with very few vertical crosslinks (Bentzel et al., 1980). Similarly, treatment with the actin-depolymerizing drug cytochalasin D, which decreases TER in guinea pig ileum, results in the transformation of the strand network from a regularly crosslinked meshwork to one that is sparse and disorganized (Madara et al., 1986). Thus, in addition to claudin composition, strand density, orientation and crosslinking may impact barrier function (Fig. 1).

Mechanical changes in junctional or tissue tension can also alter the strand network morphology by reorienting the strand network or inducing strand breaks (Fig. 2; see also Hull and Staehelin, 1976; Pitelka and Taggart, 1983). Interestingly, radial tension generated by contraction of the apical actomyosin array resulted in networks with more vertically oriented strands, whereas tension generated by tissue stretch generated more parallel TJ strands (Pitelka and Taggart, 1983). This indicates that tissue mechanics may influence strand network morphology, in turn impacting barrier function.

Fig. 2.

Potential mechanisms to elongate the strand network. These include strand straightening and reduction of interstrand crosslinks through strand breakage, reorientation and joining. In addition, network elongation may involve new strand synthesis by incorporation of claudins at free strand ends or de novo strand assembly.

Fig. 2.

Potential mechanisms to elongate the strand network. These include strand straightening and reduction of interstrand crosslinks through strand breakage, reorientation and joining. In addition, network elongation may involve new strand synthesis by incorporation of claudins at free strand ends or de novo strand assembly.

Although FFEM is considered the ‘gold standard’ tool for visualizing TJ strands because it provides high-resolution information about the morphology of the strand network, this technique presents limitations, including limited molecular specificity and dependence on tissue fixation, meaning that strand dynamics cannot be observed over time (Bartle et al., 2018). Improvements to super-resolution imaging techniques might make high-resolution live imaging of the dynamics of epithelial cell TJ strands and proteins that associate with them possible in the future.

In the meantime, expression of claudins in fibroblasts, which do not normally make TJs, has been an effective approach to reconstitute dynamic strand networks similar to those seen by FFEM in epithelial cells (Furuse et al., 1998). Because the plane of cell–cell contact in fibroblasts is parallel to the imaging plane and the strands are less densely packed than in native TJs, it is possible to resolve individual strands by using light microscopy or super-resolution microscopy (Sasaki et al., 2003; Kaufmann et al., 2012; Van Itallie et al., 2017). However, it should be noted that reconstituted claudin strands lack the full complement of TJ proteins, meaning that strand dynamics in native TJs could vary substantially. These reconstitution experiments revealed that claudin strands can be remarkably dynamic within the membrane, capable of breaking and annealing, as well as forming transient end-to-side and side-to-side interactions (Sasaki et al., 2003; Van Itallie et al., 2017). These observations led to the hypothesis that the breaking and annealing of TJ strands is the mechanistic basis that underlies the leak pathway (Sasaki et al., 2003; Van Itallie et al., 2017; Zihni et al., 2016), the non-selective flux of macromolecules across tissues (Box 2).

To better understand how interactions between claudins and other TJ proteins influence strand dynamics, the TJ proteins ZO-1 and occludin were also introduced into fibroblasts expressing claudin-2. The reconstituted strands that were linked to actin through ZO-1 aligned with actin filaments (Van Itallie et al., 2017). Although the overall size of strand patches was stabilized when they were linked to actin through ZO-1, the dynamic breaking and joining of strands was not changed, indicating that this interaction alone is not sufficient to stabilize strand breaks (Van Itallie et al., 2017). Interestingly, occludin tends to localize to free strand ends and branch points, which are more susceptible to breaking and appear to be sites of claudin incorporation (Van Itallie et al., 2017; see also Fig. 2). Because reconstituted strands are likely to behave differently than native strands, future studies that can observe TJ strands and single-molecule dynamics in vivo would be a major leap forward in our understanding of how proteins, such as ZO-1 and occludin, modify TJ dynamics, as well as how strand dynamics contribute to barrier function.

In summary, claudin-based strands form the selectively permeable seal that generates the barrier function of the TJ. Strand organization and function is regulated by multiple factors including mechanical forces that change tissue tension, connection of the transmembrane TJ proteins to the actin cytoskeleton and molecular composition of the TJ. We next discuss the dynamics of TJ proteins at the molecular scale.

Molecular dynamics

Fluorescence recovery after photobleaching (FRAP) is a powerful tool for understanding TJ remodeling at the molecular level. FRAP of claudins in individual reconstituted strands in fibroblasts revealed that, once incorporated into strands, claudins are locked in place – there is no fluorescence recovery (Sasaki et al., 2003; Van Itallie et al., 2017). Rather, newly synthesized claudin molecules are likely to be added at strand ends (Van Itallie et al., 2017).

In epithelial TJs, FRAP of claudins reveals that they, too, are remarkably stable, with typical mobile fractions between 20 and 35% (Shen et al., 2008; Yu et al., 2010; Raleigh et al., 2011; Capaldo et al., 2014; Higashi et al., 2016). Several studies suggest that claudin stability is associated with claudin function and incorporation into claudin strands. For example, claudin-2 is a pore-forming claudin that is permeable to cations. When occludin serine residue 408 is not phosphorylated, it forms a complex with ZO-1 and claudin-2, and claudin-2 mobility increases. Under these circumstances, TER increases (i.e. ion permeability decreases), indicating loss of claudin-2 pore functionality (Raleigh et al., 2011). A similar pattern is observed when Caco-2 cells are treated with the pro-inflammatory cytokines interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) (Capaldo et al., 2014). Cytokine treatment decreases TER, which is associated with increased mobility of claudin-4, a barrier-forming claudin and decreased mobility of claudin-2 (Capaldo et al., 2014). Additionally, claudins that are capable of forming reconstituted strands in the epithelial-like cell line SF7 have lower mobile fractions than other claudins that are unable to do so (Yamazaki et al., 2011). The simplest interpretation of these results is that incorporation of claudins into strands reduces their diffusion and allows them to act as either pores or diffusion barriers, according to the type of claudin.

FRAP and complementary techniques, such as fluorescence loss in photobleaching (FLIP) and photoactivation, have also provided information about the molecular dynamics of TJ proteins over short periods of time (seconds to minutes), demonstrating that FRAP recovery of occludin and claudins occurs through lateral diffusion within the membrane; although diffusion of claudins is more spatially restricted than that of occludin, and the mobile fraction of occludin is higher, between 50 and 80% (Shen et al., 2008; Yu et al., 2010; Raleigh et al., 2011; Van Itallie et al., 2015; Ronaghan et al., 2016; Janosevic et al., 2016). Over longer periods of time (hours), claudins and occludin also behave differently. A recent study using pulse labeling of SNAP- and CLIP-tagged TJ proteins has shown that occludin and claudins move within the TJ in unique ways (Van Itallie et al., 2019). Newly synthesized claudins are trafficked to the basolateral membrane and are incorporated into free strand ends on the basal side of the TJ. As strands mature, claudins move apically and old claudins are removed by endocytosis. By contrast, occludin trafficking is not polarized, and occludin can exchange between TJ and basolateral pools. This further implies that claudins are ‘locked in’ to strands, whereas occludin is more transiently associated with strands or other TJ structures (Van Itallie et al., 2019).

ZO-1 exchange occurs not through diffusion along the membrane but through exchange with a cytoplasmic pool (Shen et al., 2008; Yu et al., 2010). According to FRAP studies, ∼10–50% of ZO-1 is immobile (Shen et al., 2008; Yu et al., 2010; Raleigh et al., 2011; Van Itallie et al., 2015; Higashi et al., 2016). Stabilization at the junction requires the actin-binding region of ZO-1 and might also reflect stable association with structures, such as claudin strands. Recent evidence suggests that ZO family proteins form phase-separated compartments (Beutel et al., 2019, Schwayer et al., 2019), and many TJ proteins, including scaffolding proteins, signaling proteins and the cytoplasmic tails of TJ membrane proteins, are concentrated within these compartments – in some cases as much as 40-fold higher than the cytoplasmic concentration (Beutel et al., 2019). The dense concentration of TJ proteins within these phase-separated compartments is likely to allow for emergent TJ properties, such as the polymerization of claudins into strands, which requires ZO-1 or ZO-2 and their ability to multimerize, as well as the ability of ZO-1 to bind actin (Beutel et al., 2019; Otani et al., 2019; Schwayer et al., 2019; Umeda et al., 2006). Understanding the molecular dynamics and organization of TJ proteins will give us a stronger foundation to explain how TJ strands and molecules respond to cell- and tissue-scale dynamics.

Cell-scale dynamics

Adherens junctions (AJs) are typically viewed as the main load-bearing actin-associated cell–cell junctions, and their role in force sensing during cell shape change and development has been extensively studied (Charras and Yap, 2018). However, it is less clear how TJs respond to cell shape changes, particularly when existing cell–cell interfaces are elongated or new cell–cell interfaces are established, such as during cell division and cell extrusion (Fig. 1, cells).

During cell division, a relatively small portion of the cell–cell junction experiences direct pulling forces from the contractile ring. In Xenopus laevis embryos, FRAP of AJ proteins showed that they are stabilized at the cleavage furrow; however, FRAP does not detectably change for TJ proteins, indicating that AJs are coupled to the forces that are generated by the actomyosin contractile ring (Higashi et al., 2016). Interestingly, ZO-1 intensity at the furrow does increase relative to that at the poles, indicating that some TJ remodeling occurs during cytokinesis (Higashi et al., 2016). Indeed, FFEM showed that TJs in mitotic cells are more disorganized, comprising a higher number of free strand ends than non-mitotic cells (Tice et al., 1979).

Cell extrusion presents another potential challenge to epithelial barrier integrity. During extrusion, which occurs due to cell death or overcrowding of healthy cells in the epithelial monolayer, cells are squeezed out of epithelial tissues by their neighbors (Gudipaty and Rosenblatt, 2017). During this process, TJs extend basally to maintain contact between neighboring cells and the extruded cell (Madara, 1990; Guan et al., 2011; Marchiando et al., 2011). Following cell extrusion, new cell–cell interfaces must be formed.

During events involving cell-scale dynamics, such as cell division and cell extrusion, major shape changes occur not only in cell interfaces that are directly involved in the process but also in neighboring cells. These cells must then change their shape to accommodate the movements of the dividing or extruding cells. During both cytokinesis and extrusion, the epithelial barrier is largely maintained (Higashi et al., 2016; Rosenblatt et al., 2001; Madara, 1990; Guan et al., 2011; Marchiando et al., 2011). Therefore, gaining a better understanding of the mechanisms that allow TJs to expand and contract, while simultaneously restricting ion and macromolecule flux, is an important goal for this field of research.

Tissue-scale dynamics

Tissue-scale dynamics include TJ remodeling over extended periods of time and greater distances during development, morphogenesis and in diseased states. For example, maturation of TJs as tissues differentiate during development involves the expression of different sets of tissue-specific TJ proteins and signaling proteins (Günzel and Yu, 2013). Furthermore, tissue-scale mechanical stretching during developmental morphogenesis or organ-specific functions (e.g. bladder filling and voiding) requires significant TJ remodeling. Additionally, disease states, such as pathological inflammation, can influence the morphology, protein composition and barrier function of TJs at the tissue scale by modulating TJ protein expression and trafficking, as well as actomyosin structure (please see the excellent reviews by Luissint et al., 2016; Garcia-Hernandez et al., 2017; Marchiando et al., 2010).

During developmental morphogenesis, dynamic remodeling of TJs is not only required to maintain barrier function as epithelial cells undergo shape changes and rearrangements, but TJs also actively help to drive development. Several studies have shown that TJ proteins are crucial for apical-basal polarity, blastocyst and lumen formation, and convergent extension (Moriwaki et al., 2007; Navis and Bagnat, 2015; Baumholtz et al., 2017; Otani et al., 2019). For example, loss of ZO-1 or occludin results in multi-lumen formation in 3D epithelial culture (Odenwald et al., 2018; Otani et al., 2019). Additionally, disruption of claudins impairs expansion of the blastocoel, a fluid-filled lumen, due to a leaky barrier and loss of hydrostatic pressure (Moriwaki et al., 2007). Furthermore, TJ-dependent fluid accumulation generates the hydrostatic pressure essential for lumen formation and expansion during organogenesis, as clearly demonstrated in the zebrafish gut (Navis and Bagnat, 2015). Together, these studies suggest that TJs play a crucial role during development that extends beyond their role in regulating paracellular permeability.

Having defined a multiscale framework within which we can consider TJ remodeling, we next highlight a recent study from our lab that unifies TJ and barrier dynamics across scales.

We recently demonstrated that, when cell–cell junctions elongate in the Xenopus laevis embryonic epithelium, local discontinuities in ZO-1 and occludin, but not claudin-6, often occur along the expanding junction (Stephenson et al., 2019). By using a highly sensitive barrier assay, the zinc-based ultrasensitive microscopic barrier assay (ZnUMBA), we identified that the local breaks correspond to sites of leak pathway flux (Stephenson et al., 2019). Importantly, these leaks were quickly repaired by Rho flares, local activations of RhoA that promote local actin accumulation and myosin II-mediated contraction, which concentrates TJ proteins to reinforce the barrier at sites of local leaks (Fig. 3).

Fig. 3.

TJs are remodeled in response to local changes in tension. Elongation of a junction near a dividing cell (boxed area, magnification shown below) causes local loss of ZO-1 and a breach in barrier function (not shown). Activation of RhoA (green dome) at the site of ZO-1 loss promotes actomyosin-mediated contraction of junctions (blue arrows), which concentrates TJ proteins to repair the barrier leak. Modified with permission from Stephenson et al., 2019.

Fig. 3.

TJs are remodeled in response to local changes in tension. Elongation of a junction near a dividing cell (boxed area, magnification shown below) causes local loss of ZO-1 and a breach in barrier function (not shown). Activation of RhoA (green dome) at the site of ZO-1 loss promotes actomyosin-mediated contraction of junctions (blue arrows), which concentrates TJ proteins to repair the barrier leak. Modified with permission from Stephenson et al., 2019.

Breaking and annealing of TJ strands is thought to be the means that allows macromolecules to cross the TJ (Zihni et al., 2016). Owing to its indiscriminate nature and increased prevalence in diseased tissues, leak pathway flux could be assumed to be an ‘accidental’ feature of TJs rather than a purposeful one. We propose that the ability of TJ strands to easily break gives cells the flexibility to change their shape without being constrained by a rigid paracellular barrier. Actomyosin contraction then drives reinforcement of the TJ barrier by locally increasing protein concentration. Because reconstituted strand ends readily re-anneal with one another (Van Itallie et al., 2017), locally increasing protein concentration may facilitate this process (Fig. 1, strands and cells). Additionally, there might be yet-unknown functions of transient increases in permeability; perhaps increased flux contributes to junction remodeling or these strand breaks are necessary for transepithelial migration.

Not every instance of junction elongation causes a leak or is followed by contraction. Therefore, Rho flares might be an emergency repair mechanism that kicks in when a TJ is more severely damaged. However, this raises the question: what is the routine maintenance mechanism that allows TJs to elongate gradually while maintaining the barrier? In tissues that have been experimentally stretched, TJ strands align with the axis of tension; they appear highly taut and are reduced in number (Pitelka and Taggart, 1983; Hull and Staehelin, 1976). These features indicate that a reorientation of the strand network in response to stretch could help to cover an expanding area without de novo synthesis of TJ strands (Fig. 2). Breaks in strand networks and free strand ends are also a common feature of remodeling TJ networks (Van Itallie et al., 2019). In reconstituted TJ strands, free ends can rapidly reanneal to existing strands and are also the sites of new claudin incorporation (Van Itallie et al., 2017). Thus, strand reannealing and claudin addition may help TJ strands to cover more linear distance, ensuring that a stretched cell maintains barrier function.

In the following section, we speculate on how our knowledge about global perturbations of TJ proteins can be applied to understand TJ remodeling on local, short-term scales.

The function and remodeling of TJs have been studied by using genetic manipulations of specific TJ proteins, as well as by modulating cytoskeletal tension using small-molecule inhibitors or mechanical stretch. In this section, we focus on experiments that test the functions of ZO proteins, occludin and cytoskeletal tension in TJ establishment and barrier function.

Double knockout of ZO-1 and ZO-2 in MDCK II cells results in failure to form TJ strands and, thereby, increased flux of ions through the pore pathway and of macromolecules through the leak pathway (Otani et al., 2019). Previous knockdown studies that investigated partial loss of ZO-1 or ZO-2, or of both ZO proteins resulted in a number of less-severe phenotypes, also pointing to the importance of ZO proteins in the regulation of TJ structure and epithelial barrier function (Van Itallie et al., 2009; Fanning et al., 2012; Rodgers et al., 2013; Odenwald et al., 2018; Umeda et al., 2006). Possible reasons for variability among these studies include redundancy between ZO-1 and ZO-2, the use of different epithelial cell lines comprising different baseline levels of TJ proteins, and variations regarding the level of ZO protein knockdown. Nevertheless, common themes that emerged from these studies include that complete or partial loss of ZO proteins correlates with the robust accumulation of F-actin and myosin II, as well as an increased flux of macromolecules through the leak pathway (Van Itallie et al., 2009; Fanning et al., 2012; Rodgers et al., 2013; Odenwald et al., 2018; Umeda et al., 2006; see also Fig. 4A). Notably, this aligns with our finding that the transient TJ breaks prior to Rho flares are, indeed, associated with local reduction of ZO-1, as well as with increased flux via the leak pathway, which is soon followed by local F-actin accumulation (Stephenson et al., 2019; see also Fig. 3).

Fig. 4.

TJs are remodeled in response to tissue-scale changes in tensionover longer and shorter periods of time. (A) Sustained global remodeling of TJs through genetic manipulation – specifically ZO-1 and ZO-2 double knockdown. In epithelial cells, double knockdown of ZO-1 and ZO-2 increases the accumulation of perijunctional actomyosin, and cells exhibit increased paracellular flux of large solutes (not illustrated). A magnification of the boxed area is shown below, showing this region before (top) and after (bottom) double knockdown of ZO-1 and ZO-2. (B) Acute global remodeling of TJs through mechanical stimuli. Mechanical stretching of epithelial tissue (indicated by black arrows) also increases accumulation of perijunctional actomyosin, and cells exhibit barrier leaks (not shown). A magnification of the boxed area is shown below, showing this region before (top) and after (bottom) stretch. See text for additional details.

Fig. 4.

TJs are remodeled in response to tissue-scale changes in tensionover longer and shorter periods of time. (A) Sustained global remodeling of TJs through genetic manipulation – specifically ZO-1 and ZO-2 double knockdown. In epithelial cells, double knockdown of ZO-1 and ZO-2 increases the accumulation of perijunctional actomyosin, and cells exhibit increased paracellular flux of large solutes (not illustrated). A magnification of the boxed area is shown below, showing this region before (top) and after (bottom) double knockdown of ZO-1 and ZO-2. (B) Acute global remodeling of TJs through mechanical stimuli. Mechanical stretching of epithelial tissue (indicated by black arrows) also increases accumulation of perijunctional actomyosin, and cells exhibit barrier leaks (not shown). A magnification of the boxed area is shown below, showing this region before (top) and after (bottom) stretch. See text for additional details.

Like ZO-1 depletion, genetic perturbation of occludin is also associated with increased flux via the leak pathway (Turner et al., 2014; Buschmann et al., 2013). Interestingly, global knockdown or knockout of occludin has no observable effect on strand network morphology when using FFEM (Yu et al., 2005); thus, how exactly occludin perturbation increases flux via the leak pathway is unknown. However, the local loss of ZO-1 and occludin we observed prior to Rho flares (Stephenson et al., 2019) suggests that Rho flares have a direct role in regulating the barrier to macromolecules. For instance, it is possible that ZO-1 and occludin change the dynamics of TJ strands, thereby, modulating barrier function. However, such a change in TJ strand dynamics would not be visible by FFEM.

The function and remodeling of TJs can be altered over shorter periods of time by modulating actomyosin cytoskeletal tension. Acute treatments that inhibit actin polymerization lead to increased paracellular permeability due to mislocalization of TJ proteins, including ZO-1 and occludin (Van Itallie et al., 2009, 2015; Fanning et al., 2012; Shen and Turner, 2005). Further, inhibition of myosin II activity mediated by Rho kinase (ROCK) or myosin light chain kinase (MLCK) alters the contractility of the apical actomyosin bundle, thus, affecting the permeability of TJs (Nusrat et al., 1995; Clayburgh et al., 2004; Walsh et al., 2001). For example, inhibition of ROCK results in increased flux of macromolecules and ions (Walsh et al., 2001). By contrast, inhibition of MLCK stabilizes ZO-1 at TJs by coupling it to the apical actomyosin array, thereby, restricting the flux of ions (Yu et al., 2010). Taken together, these results suggest that, although ROCK and MLCK both regulate myosin II activity through phosphorylation of myosin II regulatory light chain, these kinases have different effects on TJ barrier function. Interestingly, the local reductions in ZO-1 and occludin we detected prior to Rho flares are followed by accumulation of F-actin and myosin II (Fig. 3), and ROCK-mediated myosin II activity is required for efficient reinstatement of ZO-1 at the sites of barrier leaks (Stephenson et al., 2019).

Mechanical forces change tissue tension and promote associated changes in the actin cytoskeleton and TJ remodeling at a global scale. For instance, when alveolar epithelial cells are stretched biaxially in a cyclic manner, they exhibit an increase in paracellular permeability and in perijunctional actin levels (Cavanaugh et al., 2001; DiPaolo and Margulies, 2012). Additionally, upon biaxial stretch, the overall levels of ZO-1 and occludin at TJs are reduced, and regions of alternating high and low TJ protein intensities are visible along the junction (Cavanaugh et al., 2001). A similar phenotype, i.e. loss of ZO-1 and occludin at TJs, was observed in biaxially stretched Caco-2 cells (Samak et al., 2014; see also Fig. 4B). These studies suggest that stretch-induced cytoskeletal rearrangements perturb the uniform, continuous localization of TJ proteins along cell–cell junctions, thus disrupting barrier function. However, it remains unclear whether (i) the observed discontinuities in ZO-1 and occludin, indeed, correspond to local barrier breaches and (ii) the discontinuities in ZO-1 and occludin are cause or consequence of stretch-induced cytoskeletal rearrangement. To this end, Samak et al. have shown that inhibition of MLCK prior to experimental tissue stretch attenuated the loss of ZO-1 and occludin, suggesting that MLCK-mediated cytoskeletal rearrangement promotes discontinuities of TJs (Samak et al., 2014). Together, these results highlight the importance of feedback regulation between the actomyosin cytoskeleton and TJ proteins, as TJ proteins respond to actomyosin-mediated mechanical forces, and partial or complete loss of ZO proteins leads to robust accumulation of F-actin and myosin II. However, further studies are required to determine the mechanistic nature of this feedback.

So far, we have discussed multiple temporal and spatial scales at which TJs are dynamic, and our current understanding of some of the mechanisms that drive TJ remodeling at global and local scales. We conclude by highlighting two emerging areas in TJ research, mechanotransduction and super-resolution imaging, and outline how future work may enhance our understanding of mechanisms that drive TJ remodeling.

Mechanotransduction at the tight junction

Mechanotransduction is a process by which cells sense mechanical force and respond by transmitting biochemical signals through cellular signaling pathways. Mechanotransduction at AJs is evident in response to both tissue-scale and locally applied forces (Charras and Yap, 2018; Pinheiro and Bellaïche, 2018). Tensile forces, for example, can change the conformation of mechanosensitive AJ proteins, resulting in the recruitment of additional proteins to reinforce and stabilize junctions (Yonemura et al., 2010; Le Duc et al., 2010; Liu et al., 2010). However, much less is known about mechanotransduction at TJs (Sluysmans et al., 2017).

A recent study identified a mechanosensitive pathway that maintains epithelial integrity and barrier function through activation of Rho (Acharya et al., 2018). In that study, the mechanical stretching of cultured epithelial cells activates Rho through the recruitment of a specific Rho guanine nucleotide exchange factor (RhoGEF), p114 RhoGEF. Active Rho then activates the formin mDia1 (also known as Diaph1) to promote actin assembly, which, in turn, strengthens the junctions (Fig. 4B; see also Acharya et al., 2018). Of note, p114 RhoGEF also promotes Rho activation during TJ assembly (Terry et al., 2011; Xu et al., 2013), suggesting that this mechanosensitive pathway may also be at play at TJs.

Seminal EM studies carried out in epithelial chick hair cells identified two distinct, yet interconnected, actin populations at junctions: (i) bundles of actin filaments that run parallel to the membrane at the AJ and, (ii) a meshwork of filaments that lies just beneath the TJ (Hirokawa, 1982; Hirokawa et al., 1983). Indeed, these EM data underlie most of the currently held hypotheses about connections of actin to TJs and AJs, leading us to assume that transmission of tension primarily takes place at the AJ. However, more evidence is needed to determine whether AJs are, indeed, the primary load-bearing actin-associated cell–cell junction or whether TJs can also be load-bearing structures.

Several pieces of data suggest that reassessment of our current understanding of actin connections to the TJ and AJ is warranted. First, we recently quantified the apico-basal localization of actin with respect to the TJ and AJ in the Xenopus embryonic epithelium, and found that TJ-associated F-actin is more abundant than AJ-associated F-actin in these cells (Higashi et al., 2019). Second, tricellular junctions, the points where three cells come together, may utilize specialized actin connections (see the recent review by Higashi and Miller, 2017). Recent studies have suggested that F-actin makes end-on (versus side-on) connections at tricellular junctions (Yonemura, 2011; Choi et al., 2016). We propose that these end-on actin connections are important for tension transmission and barrier function at tricellular junctions, and recruit different protein partners than side-on actin connections to offset the increased force applied on tricellular junctions (Higashi and Miller, 2017). Third, by using platinum replica EM, a recent examination of connections between actin and junctions in vascular endothelial cells revealed that AJs in these cells connect to Arp2/3-containing branched actin networks but to not bundled actomyosin (Efimova and Svitkina, 2018), raising the question of whether connections to branched actin networks are also important in polarized epithelial cells. Fourth, together, these studies suggest that actin connections to apical cell–cell junctions can differ from the commonly held model of a perijunctional actomyosin ring that runs parallel to the AJ. Future studies should directly investigate whether TJs are load-bearing structures. To this end, the use of tension biosensors – similar to those developed to measure mechanical stresses at focal adhesions and AJs at the molecular level (Grashoff et al., 2010; Hoffman and Yap, 2015) – but, instead, designed to measure tensile force applied on specific TJ proteins, would be immensely useful.

By applying our knowledge of mechanotransduction at AJs to that of TJs, it seems possible that TJ proteins also undergo tension-dependent conformational changes analogous to the conformational changes exhibited by AJ proteins, such as α-catenin (Yonemura et al., 2010), and/or that specific proteins are recruited to the TJ under increased tension. Indeed, ZO-1 has recently been shown to undergo a tension-dependent conformational change, opening to a stretched conformation in response to actomyosin-generated force (Spadaro et al., 2017). Here, unfolding of ZO-1 occurs at a level of force similar to that needed to unfold α-catenin in AJs (>5 pN for ZO-1 compared with 5–15 pN for α-catenin) (Spadaro et al., 2017; Yao et al., 2014). This conformational change allows ZO-1 to recruit occludin and a transcription factor to TJs (Spadaro et al., 2017). Supporting the conclusion that ZO-1 is a mechanosensitive protein, FRAP experiments have shown that ZO-1 is stabilized in Xenopus embryos under increased tension (Higashi et al., 2016), suggesting that TJs can, indeed, sense increased tension and respond by stabilizing ZO-1.

What are the potential functional effects of mechanotransduction at TJs? Several pieces of evidence suggest that mechanosensitive pathways are important for maintenance of barrier function under tensile stress. Proteins that are known to be recruited to cell–cell junctions in response to tension, such as afadin and vinculin, are necessary to maintain tissue integrity and barrier function under high tension (Choi et al., 2016; Le Duc et al., 2010). Furthermore, a recently identified mechanosensitive pathway helps to maintain epithelial integrity and barrier function by activating Rho at junctions (Acharya et al., 2018). In the future, it will be interesting to further explore mechanotransduction pathways in epithelial tissues that exhibit cyclic stretch and release, such as filling and emptying of the bladder or the lung (Carattino et al., 2013), or gut peristalsis (Trepat et al., 2007), as in these tissues, the ability to sense and respond to force is likely to be essential for barrier maintenance. Additionally, mechanotransduction at TJs was recently shown to play a role in epithelial tissue spreading during zebrafish gastrulation (Schwayer et al., 2019). In that study, accumulation of TJ components, not AJ components, scaled with the tension of the adjacent actomyosin network, indicating that that these TJs are mechanosensitive. Transportation of phase-separated non-junctional ZO-1 clusters towards the TJ by actomyosin-driven flow was required for the mechanosensitivity of these TJs. This study opens the door for future studies of functional roles for TJs in mediating mechanotransduction during developmental or homeostatic processes. Finally, because mechanosensitive junction proteins can also be activated to undergo conformational changes and recruit other proteins in response to locally applied forces, such as the cytokinetic contractile ring (Higashi et al., 2016), it will be interesting to explore whether local TJ remodeling events involve mechanotransduction and whether they are important for barrier function.

Super-resolution microscopy of TJ organization and dynamics

Traditionally, EM techniques, including transmission electron microscopy (TEM) and FFEM, have been used to obtain an ultrastructural view of TJs. Indeed, these approaches have defined our understanding of the structural organization of TJs (e.g. Farquhar and Palade, 1963; Chalcroft and Bullivant, 1970). However, EM cannot capture dynamic changes in TJ strands or the associated cytoskeleton, which underlie the dynamic regulation of barrier function. Light-microscopy techniques, by contrast, are able to reveal the dynamic nature of TJs but, due to the limit the diffraction of light imposes on spatial resolution, these approaches are limited to a resolution of ∼250 nm (under optimal conditions) (Bartle et al., 2018; Schermelleh et al., 2019). Because the size of TJs and other epithelial cell–cell junctions are near this resolution limit (Bartle et al., 2018), the information one can obtain about TJ composition, structure and dynamics is restricted when using light microscopy.

Super-resolution microscopy can bridge the gap between light microscopy and EM. By using different strategies to measure just a subset of fluorophores in the sample, super-resolution microscopy techniques – including stimulated emission depletion (STED) and structural illumination microscopy (SIM), as well as single-molecule localization microscopy techniques, such as fluorescence photoactivated localization microscopy (F-PALM) or stochastic optical reconstruction microscopy (STORM) – overcome the resolution limit of light microscopy, leading to an improved resolution in the range of 20–100 nm (Bartle et al., 2018; Schermelleh et al., 2019; Lambert and Waters, 2017). These techniques combine molecular specificity (i.e. labeling proteins of interest) with high-resolution spatial information. Additionally, they provide an advantage over EM for imaging the actin cytoskeleton associated with TJs, because cells do not have to be treated with harsh fixatives, which often disrupt the cytoskeleton. Recently, several studies have applied super-resolution microscopy techniques to study TJ proteins, and some of these studies reveal novel information about TJ remodeling (Table S1).

The emergence of more widely accessible super-resolution microscopy techniques opens new avenues in TJ research and is expected to reveal new insights into the structure and dynamic remodeling of TJs. There are several aspects of barrier biology, in particular, that could benefit from super-resolution imaging: (i) determining the nanoscale organization of claudins and occludin in strand networks in reconstituted systems, epithelial cells, organoids, and/or developing model organisms; (ii) investigating the mechanisms of delivery and incorporation of TJ proteins into sealing strands, as well as TJ protein turnover; (iii) investigating remodeling of TJs during cytokinesis to understand how barrier function is maintained during cell division and; (iv) determining how the actin cytoskeleton is connected to TJs and tricellular junctions, and how these connections are remodeled under different tension conditions.

In this Review, we have highlighted the dynamic remodeling of TJs to maintain an effective barrier in response to cell- and tissue-scale forces. We argue that, in order to understand TJ remodeling, we must investigate TJ remodeling at multiple scales. Although the functional roles of many TJ proteins in maintaining an intact barrier have been characterized on an individual basis, it is largely unknown how TJ strands and strand networks are dynamically remodeled in response to physiological or pathological stimuli. To address these questions, we suggest several areas of interest for further investigation. First, understanding the kinetics of claudin strand assembly, will help to answer the question of whether claudins self-assemble or whether other proteins facilitate strand formation. Second, additional structural information about claudins and occludin, including their binding interfaces, will shed light on how strands branch, break and anneal, and might help reveal the exact function of occludin. Finally, observing the dynamics of TJ strand remodeling in epithelial cells will determine how strand networks are dynamically remodeled in vivo.

We thank members of the Miller lab for useful discussion and feedback, Tomohito Higashi for providing critical feedback on a draft of this Review, and the three anonymous referees for their insightful comments and suggestions.

Funding

Research in A.L.M.’s lab is supported by grants from the National Institutes of Health (NIH; grant number: R01 GM112794) and the National Science Foundation (NSF; award number: 1615338). Deposited in PMC for release after 12 months.

Acharya
,
B. R.
,
Nestor-Bergmann
,
A.
,
Liang
,
X.
,
Gupta
,
S.
,
Duszyc
,
K.
,
Gauquelin
,
E.
,
Gomez
,
G. A.
,
Budnar
,
S.
,
Marcq
,
P.
,
Jensen
,
O. E.
, et al. 
(
2018
).
A mechanosensitive RhoA pathway that protects epithelia against acute tensile stress
.
Dev. Cell
47
,
439
-
452.e6
.
Anderson
,
J. M.
and
Van Itallie
,
C. M.
(
2009
).
Physiology and function of the tight junction
.
Cold Spring Harb. Perspect Biol.
1
,
a002584
.
Arnold
,
T. R.
,
Stephenson
,
R. E.
and
Miller
,
A. L.
(
2017
).
Rho GTPases and actomyosin: partners in regulating epithelial cell-cell junction structure and function
.
Exp. Cell Res.
358
,
20
-
30
.
Balda
,
M. S.
,
Whitney
,
J. A.
,
Flores
,
C.
,
Gonzalez
,
S.
,
Cereijido
,
M.
and
Matter
,
K.
(
1996
).
Functional dissociation of paracellular permeability and transepithelial electrical resistance and disruption of the apical-basolateral intramembrane diffusion barrier by expression of a mutant tight junction membrane protein
.
J. Cell Biol.
134
,
1031
-
1049
.
Bartle
,
E. I.
,
Rao
,
T. C.
,
Urner
,
T. M.
and
Mattheyses
,
A. L.
(
2018
).
Bridging the gap: Super-resolution microscopy of epithelial cell junctions
.
Tissue Barriers
6
,
e1404189
.
Baumholtz
,
A. I.
,
Simard
,
A.
,
Nikolopoulou
,
E.
,
Oosenbrug
,
M.
,
Collins
,
M. M.
,
Piontek
,
A.
,
Krause
,
G.
,
Piontek
,
J.
,
Greene
,
N. D. E.
and
Ryan
,
A. K.
(
2017
).
Claudins are essential for cell shape changes and convergent extension movements during neural tube closure
.
Dev. Biol.
428
,
25
-
38
.
Bentzel
,
C. J.
,
Hainau
,
B.
,
Edelman
,
A.
,
Anagnostopoulos
,
T.
and
Benedetti
,
E. L.
(
1976
).
Effect of plant cytokinins on microfilaments and tight junction permeability
.
Nature
264
,
666
-
668
.
Bentzel
,
C. J.
,
Hainau
,
B.
,
Ho
,
S.
,
Hui
,
S. W.
,
Edelman
,
A.
,
Anagnostopoulos
,
T.
and
Benedetti
,
E. L.
(
1980
).
Am. J. Physiol.
239
,
C75
-
C89
.
Beutel
,
O.
,
Maraspini
,
R.
,
Pombo-Garcia
,
K.
,
Martin-Lemaitre
,
C.
and
Honigmann
,
A.
(
2019
).
Phase separation of zonula occludens proteins drives formation of tight junctions
.
Cell
179
,
923
-
936
. e11.
Buschmann
,
M. M.
,
Shen
,
L.
,
Rajapakse
,
H.
,
Raleigh
,
D. R.
,
Wang
,
Y.
,
Wang
,
Y.
,
Lingaraju
,
A.
,
Zha
,
J.
,
Abbott
,
E.
,
Mcauley
,
E. M.
, et al. 
(
2013
).
Occludin OCEL-domain interactions are required for maintenance and regulation of the tight junction barrier to macromolecular flux
.
Mol. Biol. Cell
24
,
3056
-
3068
.
Capaldo
,
C. T.
,
Farkas
,
A. E.
,
Hilgarth
,
R. S.
,
Krug
,
S. M.
,
Wolf
,
M. F.
,
Benedik
,
J. K.
,
Fromm
,
M.
,
Koval
,
M.
,
Parkos
,
C.
and
Nusrat
,
A.
(
2014
).
Mol. Biol. Cell
25
,
2710
-
2719
.
Carattino
,
M. D.
,
Prakasam
,
H. S.
,
Ruiz
,
W. G.
,
Clayton
,
D. R.
,
Mcguire
,
M.
,
Gallo
,
L. I.
and
Apodaca
,
G.
(
2013
).
Bladder filling and voiding affect umbrella cell tight junction organization and function
.
Am. J. Physiol. Renal. Physiol.
305
,
F1158
-
F1168
.
Cavanaugh
,
K. J.
, Jr
.,
Oswari
,
J.
and
Margulies
,
S. S.
(
2001
).
Role of stretch on tight junction structure in alveolar epithelial cells
.
Am. J. Respir. Cell Mol. Biol.
25
,
584
-
591
.
Chalcroft
,
J. P.
and
Bullivant
,
S.
(
1970
).
An interpretation of liver cell membrane and junction structure based on observation of freeze-fracture replicas of both sides of the fracture
.
J. Cell Biol.
47
,
49
-
60
.
Charras
,
G.
and
Yap
,
A. S.
(
2018
).
Tensile forces and mechanotransduction at cell-cell junctions
.
Curr. Biol.
28
,
R445
-
R457
.
Choi
,
W.
,
Acharya
,
B. R.
,
Peyret
,
G.
,
Fardin
,
M. A.
,
Mège
,
R.-M.
,
Ladoux
,
B.
,
Yap
,
A. S.
,
Fanning
,
A. S.
and
Peifer
,
M.
(
2016
).
Remodeling the zonula adherens in response to tension and the role of afadin in this response
.
J. Cell Biol.
213
,
243
-
260
.
Claude
,
P.
and
Goodenough
,
D. A.
(
1973
).
Fracture faces of zonulae occludentes from “tight” and “leaky” epithelia
.
J. Cell Biol.
58
,
390
-
400
.
Clayburgh
,
D. R.
,
Rosen
,
S.
,
Witkowski
,
E. D.
,
Wang
,
F.
,
Blair
,
S.
,
Dudek
,
S.
,
Garcia
,
J. G.
,
Alverdy
,
J. C.
and
Turner
,
J. R.
(
2004
).
A differentiation-dependent splice variant of myosin light chain kinase, MLCK1, regulates epithelial tight junction permeability
.
J. Biol. Chem.
279
,
55506
-
55513
.
Colegio
,
O. R.
,
Van Itallie
,
C.
,
Rahner
,
C.
and
Anderson
,
J. M.
(
2003
).
Claudin extracellular domains determine paracellular charge selectivity and resistance but not tight junction fibril architecture
.
Am. J. Physiol. Cell Physiol.
284
,
C1346
-
C1354
.
Cording
,
J.
,
Berg
,
J.
,
Kading
,
N.
,
Bellmann
,
C.
,
Tscheik
,
C.
,
Westphal
,
J. K.
,
Milatz
,
S.
,
Günzel
,
D.
,
Wolburg
,
H.
,
Piontek
,
J.
, et al. 
(
2013
).
In tight junctions, claudins regulate the interactions between occludin, tricellulin and marvelD3, which, inversely, modulate claudin oligomerization
.
J. Cell Sci.
126
,
554
-
564
.
Dipaolo
,
B. C.
and
Margulies
,
S. S.
(
2012
).
Rho kinase signaling pathways during stretch in primary alveolar epithelia
.
Am. J. Physiol. Lung Cell. Mol. Physiol.
302
,
L992
-
L1002
.
Efimova
,
N.
and
Svitkina
,
T. M.
(
2018
).
Branched actin networks push against each other at adherens junctions to maintain cell-cell adhesion
.
J. Cell Biol.
217
,
1827
-
1845
.
Fanning
,
A. S.
,
Van Itallie
,
C. M.
and
Anderson
,
J. M.
(
2012
).
Zonula occludens-1 and -2 regulate apical cell structure and the zonula adherens cytoskeleton in polarized epithelia
.
Mol. Biol. Cell
23
,
577
-
590
.
Farquhar
,
M. G.
and
Palade
,
G. E.
(
1963
).
Junctional complexes in various epithelia
.
J. Cell Biol.
17
,
375
-
412
.
Furuse
,
M.
,
Sasaki
,
H.
,
Fujimoto
,
K.
and
Tsukita
,
S.
(
1998
).
A single gene product, claudin-1 or −2, reconstitutes tight junction strands and recruits occludin in fibroblasts
.
J. Cell Biol.
143
,
391
-
401
.
Garcia-Hernandez
,
V.
,
Quiros
,
M.
and
Nusrat
,
A.
(
2017
).
Intestinal epithelial claudins: expression and regulation in homeostasis and inflammation
.
Ann. N. Y. Acad. Sci.
1397
,
66
-
79
.
Grashoff
,
C.
,
Hoffman
,
B. D.
,
Brenner
,
M. D.
,
Zhou
,
R.
,
Parsons
,
M.
,
Yang
,
M. T.
,
Mclean
,
M. A.
,
Sligar
,
S. G.
,
Chen
,
C. S.
,
Ha
,
T.
, et al. 
(
2010
).
Measuring mechanical tension across vinculin reveals regulation of focal adhesion dynamics
.
Nature
466
,
263
-
266
.
Guan
,
Y.
,
Watson
,
A. J.
,
Marchiando
,
A. M.
,
Bradford
,
E.
,
Shen
,
L.
,
Turner
,
J. R.
and
Montrose
,
M. H.
(
2011
).
Redistribution of the tight junction protein ZO-1 during physiological shedding of mouse intestinal epithelial cells
.
Am. J. Physiol. Cell Physiol.
300
,
C1404
-
C1414
.
Gudipaty
,
S. A.
and
Rosenblatt
,
J.
(
2017
).
Epithelial cell extrusion: pathways and pathologies
.
Semin. Cell Dev. Biol.
67
,
132
-
140
.
Günzel
,
D.
(
2017
).
Claudins: vital partners in transcellular and paracellular transport coupling
.
Pflugers Arch.
469
,
35
-
44
.
Günzel
,
D.
and
Yu
,
A. S.
(
2013
).
Claudins and the modulation of tight junction permeability
.
Physiol. Rev.
93
,
525
-
569
.
Harris
,
T. J. C.
and
Tepass
,
U.
(
2010
).
Adherens junctions: from molecules to morphogenesis
.
Nat. Rev. Mol. Cell Biol.
11
,
502
-
514
.
Higashi
,
T.
and
Miller
,
A. L.
(
2017
).
Tricellular junctions: how to build junctions at the TRICkiest points of epithelial cells
.
Mol. Biol. Cell
.
Higashi
,
T.
,
Arnold
,
T. R.
,
Stephenson
,
R. E.
,
Dinshaw
,
K. M.
and
Miller
,
A. L.
(
2016
).
Maintenance of the epithelial barrier and remodeling of cell-cell junctions during Cytokinesis
.
Curr. Biol.
26
,
1829
-
1842
.
Higashi
,
T.
,
Stephenson
,
R. E.
and
Miller
,
A. L.
(
2019
).
Comprehensive analysis of formin localization in Xenopus epithelial cells
.
Mol. Biol. Cell
30
,
82
-
95
.
Hirokawa
,
N.
(
1982
).
Organization of actin, myosin, and intermediate filaments in the brush border of intestinal epithelial cells
.
J. Cell Biol.
94
,
425
-
443
.
Hirokawa
,
N.
,
Keller
,
T. C.
, III
,
Chasan
,
R.
and
Mooseker
,
M. S.
(
1983
).
Mechanism of brush border contractility studied by the quick-freeze, deep-etch method
.
J. Cell Biol.
96
,
1325
-
1336
.
Hoffman
,
B. D.
and
Yap
,
A. S.
(
2015
).
Towards a dynamic understanding of cadherin-based mechanobiology
.
Trends Cell Biol.
25
,
803
-
814
.
Hull
,
B. E.
and
Staehelin
,
L. A.
(
1976
).
Functional significance of the variations in the geometrical organization of tight junction networks
.
J. Cell Biol.
68
,
688
-
704
.
Janosevic
,
D.
,
Axis
,
J.
,
Bacallao
,
R. L.
and
Amsler
,
K.
(
2016
).
Occludin content modulates hydrogen peroxide-induced increase in renal epithelial paracellular permeability
.
J. Cell. Biochem.
117
,
769
-
779
.
Kaufmann
,
R.
,
Piontek
,
J.
,
Grull
,
F.
,
Kirchgessner
,
M.
,
Rossa
,
J.
,
Wolburg
,
H.
,
Blasig
,
I. E.
and
Cremer
,
C.
(
2012
).
Visualization and quantitative analysis of reconstituted tight junctions using localization microscopy
.
PLoS ONE
7
,
e31128
.
Krug
,
S. M.
,
Amasheh
,
S.
,
Richter
,
J. F.
,
Milatz
,
S.
,
Günzel
,
D.
,
Westphal
,
J. K.
,
Huber
,
O.
,
Schulzke
,
J. D.
and
Fromm
,
M.
(
2009
).
Tricellulin forms a barrier to macromolecules in tricellular tight junctions without affecting ion permeability
.
Mol. Biol. Cell
20
,
3713
-
3724
.
Lambert
,
T. J.
and
Waters
,
J. C.
(
2017
).
Navigating challenges in the application of superresolution microscopy
.
J. Cell Biol.
216
,
53
-
63
.
Le Duc
,
Q.
,
Shi
,
Q.
,
Blonk
,
I.
,
Sonnenberg
,
A.
,
Wang
,
N.
,
Leckband
,
D.
and
De Rooij
,
J.
(
2010
).
Vinculin potentiates E-cadherin mechanosensing and is recruited to actin-anchored sites within adherens junctions in a myosin II-dependent manner
.
J. Cell Biol.
189
,
1107
-
1115
.
Liu
,
Z.
,
Tan
,
J. L.
,
Cohen
,
D. M.
,
Yang
,
M. T.
,
Sniadecki
,
N. J.
,
Ruiz
,
S. A.
,
Nelson
,
C. M.
and
Chen
,
C. S.
(
2010
).
Mechanical tugging force regulates the size of cell-cell junctions
.
Proc. Natl. Acad. Sci. USA
107
,
9944
-
9949
.
Luissint
,
A. C.
,
Parkos
,
C. A.
and
Nusrat
,
A.
(
2016
).
Inflammation and the intestinal barrier: leukocyte-epithelial cell interactions, cell junction remodeling, and mucosal repair
.
Gastroenterology
151
,
616
-
632
.
Madara
,
J. L.
(
1990
).
Maintenance of the macromolecular barrier at cell extrusion sites in intestinal epithelium: physiological rearrangement of tight junctions
.
J. Membr. Biol.
116
,
177
-
184
.
Madara
,
J. L.
,
Barenberg
,
D.
and
Carlson
,
S.
(
1986
).
Effects of cytochalasin D on occluding junctions of intestinal absorptive cells: further evidence that the cytoskeleton may influence paracellular permeability and junctional charge selectivity
.
J. Cell Biol.
102
,
2125
-
2136
.
Marchiando
,
A. M.
,
Graham
,
W. V.
and
Turner
,
J. R.
(
2010
).
Epithelial barriers in homeostasis and disease
.
Annu. Rev. Pathol.
5
,
119
-
144
.
Marchiando
,
A. M.
,
Shen
,
L.
,
Graham
,
W. V.
,
Edelblum
,
K. L.
,
Duckworth
,
C. A.
,
Guan
,
Y.
,
Montrose
,
M. H.
,
Turner
,
J. R.
and
Watson
,
A. J.
(
2011
).
The epithelial barrier is maintained by in vivo tight junction expansion during pathologic intestinal epithelial shedding
.
Gastroenterology
140
,
1208
-
1218.e1-2
.
Moriwaki
,
K.
,
Tsukita
,
S.
and
Furuse
,
M.
(
2007
).
Tight junctions containing claudin 4 and 6 are essential for blastocyst formation in preimplantation mouse embryos
.
Dev. Biol.
312
,
509
-
522
.
Navis
,
A.
and
Bagnat
,
M.
(
2015
).
Developing pressures: fluid forces driving morphogenesis
.
Curr. Opin. Genet. Dev.
32
,
24
-
30
.
Nusrat
,
A.
,
Giry
,
M.
,
Turner
,
J. R.
,
Colgan
,
S. P.
,
Parkos
,
C. A.
,
Carnes
,
D.
,
Lemichez
,
E.
,
Boquet
,
P.
and
Madara
,
J. L.
(
1995
).
Rho protein regulates tight junctions and perijunctional actin organization in polarized epithelia
.
Proc. Natl. Acad. Sci. USA
92
,
10629
-
10633
.
Odenwald
,
M. A.
,
Choi
,
W.
,
Kuo
,
W. T.
,
Singh
,
G.
,
Sailer
,
A.
,
Wang
,
Y.
,
Shen
,
L.
,
Fanning
,
A. S.
and
Turner
,
J. R.
(
2018
).
The scaffolding protein ZO-1 coordinates actomyosin and epithelial apical specializations in vitro and in vivo
.
J. Biol. Chem.
293
,
17317
-
17335
.
Otani
,
T.
,
Nguyen
,
T. P.
,
Tokuda
,
S.
,
Sugihara
,
K.
,
Sugawara
,
T.
,
Furuse
,
K.
,
Miura
,
T.
,
Ebnet
,
K.
and
Furuse
,
M.
(
2019
).
Claudins and JAM-A coordinately regulate tight junction formation and epithelial polarity
.
J. Cell Biol.
218
,
3372
-
3396
.
Pinheiro
,
D.
and
Bellaïche
,
Y.
(
2018
).
Mechanical force-driven adherens junction remodeling and epithelial dynamics
.
Dev. Cell
47
,
391
.
Pitelka
,
D. R.
and
Taggart
,
B. N.
(
1983
).
Mechanical tension induces lateral movement of intramembrane components of the tight junction: studies on mouse mammary cells in culture
.
J. Cell Biol.
96
,
606
-
612
.
Raleigh
,
D. R.
,
Marchiando
,
A. M.
,
Zhang
,
Y.
,
Shen
,
L.
,
Sasaki
,
H.
,
Wang
,
Y.
,
Long
,
M.
and
Turner
,
J. R.
(
2010
).
Tight junction-associated MARVEL proteins marveld3, tricellulin, and occludin have distinct but overlapping functions
.
Mol. Biol. Cell
21
,
1200
-
1213
.
Raleigh
,
D. R.
,
Boe
,
D. M.
,
Yu
,
D.
,
Weber
,
C. R.
,
Marchiando
,
A. M.
,
Bradford
,
E. M.
,
Wang
,
Y.
,
Wu
,
L.
,
Schneeberger
,
E. E.
,
Shen
,
L.
, et al. 
(
2011
).
Occludin S408 phosphorylation regulates tight junction protein interactions and barrier function
.
J. Cell Biol.
193
,
565
-
582
.
Richter
,
J. F.
,
Schmauder
,
R.
,
Krug
,
S. M.
,
Gebert
,
A.
and
Schumann
,
M.
(
2016
).
A novel method for imaging sites of paracellular passage of macromolecules in epithelial sheets
.
J. Control. Release
229
,
70
-
79
.
Rodgers
,
L. S.
,
Beam
,
M. T.
,
Anderson
,
J. M.
and
Fanning
,
A. S.
(
2013
).
Epithelial barrier assembly requires coordinated activity of multiple domains of the tight junction protein ZO-1
.
J. Cell Sci.
126
,
1565
-
1575
.
Ronaghan
,
N. J.
,
Shang
,
J.
,
Iablokov
,
V.
,
Zaheer
,
R.
,
Colarusso
,
P.
,
Dion
,
S.
,
Désilets
,
A.
,
Leduc
,
R.
,
Turner
,
J. R.
and
Macnaughton
,
W. K.
(
2016
).
The serine protease-mediated increase in intestinal epithelial barrier function is dependent on occludin and requires an intact tight junction
.
Am. J. Physiol. Gastrointest. Liver Physiol.
311
,
G466
-
G479
.
Rosenblatt
,
J.
,
Raff
,
M. C.
and
Cramer
,
L. P.
(
2001
).
An epithelial cell destined for apoptosis signals its neighbors to extrude it by an actin- and myosin-dependent mechanism
.
Curr. Biol.
11
,
1847
-
1857
.
Samak
,
G.
,
Gangwar
,
R.
,
Crosby
,
L. M.
,
Desai
,
L. P.
,
Wilhelm
,
K.
,
Waters
,
C. M.
and
Rao
,
R.
(
2014
).
Cyclic stretch disrupts apical junctional complexes in Caco-2 cell monolayers by a JNK-2-, c-Src-, and MLCK-dependent mechanism
.
Am. J. Physiol. Gastrointest. Liver Physiol.
306
,
G947
-
G958
.
Sasaki
,
H.
,
Matsui
,
C.
,
Furuse
,
K.
,
Mimori-Kiyosue
,
Y.
,
Furuse
,
M.
and
Tsukita
,
S.
(
2003
).
Dynamic behavior of paired claudin strands within apposing plasma membranes
.
Proc. Natl. Acad. Sci. USA
100
,
3971
-
3976
.
Schermelleh
,
L.
,
Ferrand
,
A.
,
Huser
,
T.
,
Eggeling
,
C.
,
Sauer
,
M.
,
Biehlmaier
,
O.
and
Drummen
,
G. P. C.
(
2019
).
Super-resolution microscopy demystified
.
Nat. Cell Biol.
21
,
72
-
84
.
Schwayer
,
C.
,
Shamipour
,
S.
,
Pranjic-Ferscha
,
K.
,
Schauer
,
A.
,
Balda
,
M.
,
Tada
,
M.
,
Matter
,
K.
and
Heisenberg
,
C. P.
(
2019
).
Mechanosensation of Tight Junctions Depends on ZO-1 Phase Separation and Flow
.
Cell
179
,
937
-
952
. e18.
Shen
,
L.
and
Turner
,
J. R.
(
2005
).
Actin depolymerization disrupts tight junctions via caveolae-mediated endocytosis
.
Mol. Biol. Cell
16
,
3919
-
3936
.
Shen
,
L.
,
Weber
,
C. R.
and
Turner
,
J. R.
(
2008
).
The tight junction protein complex undergoes rapid and continuous molecular remodeling at steady state
.
J. Cell Biol.
181
,
683
-
695
.
Shen
,
L.
,
Weber
,
C. R.
,
Raleigh
,
D. R.
,
Yu
,
D.
and
Turner
,
J. R.
(
2011
).
Tight junction pore and leak pathways: a dynamic duo
.
Annu. Rev. Physiol.
73
,
283
-
309
.
Sluysmans
,
S.
,
Vasileva
,
E.
,
Spadaro
,
D.
,
Shah
,
J.
,
Rouaud
,
F.
and
Citi
,
S.
(
2017
).
The role of apical cell-cell junctions and associated cytoskeleton in mechanotransduction
.
Biol. Cell
109
,
139
-
161
.
Spadaro
,
D.
,
Le
,
S.
,
Laroche
,
T.
,
Mean
,
I.
,
Jond
,
L.
,
Yan
,
J.
and
Citi
,
S.
(
2017
).
Tension-dependent stretching activates ZO-1 to control the junctional localization of its interactors
.
Curr. Biol.
27
,
3783
-
3795.e8
.
Srinivasan
,
B.
,
Kolli
,
A. R.
,
Esch
,
M. B.
,
Abaci
,
H. E.
,
Shuler
,
M. L.
and
Hickman
,
J. J.
(
2015
).
TEER measurement techniques for in vitro barrier model systems
.
J. Lab. Autom.
20
,
107
-
126
.
Stephenson
,
R. E.
,
Higashi
,
T.
,
Erofeev
,
I. S.
,
Arnold
,
T. R.
,
Leda
,
M.
,
Goryachev
,
A. B.
and
Miller
,
A. L.
(
2019
).
Rho flares repair local tight junction leaks
.
Dev. Cell
48
,
445
-
459.e5
.
Terry
,
S. J.
,
Zihni
,
C.
,
Elbediwy
,
A.
,
Vitiello
,
E.
,
Leefa Chong San
,
I. V.
,
Balda
,
M. S.
and
Matter
,
K
. (
2011
).
Spatially restricted activation of RhoA signalling at epithelial junctions by p114RhoGEF drives junction formation and morphogenesis
.
Nat. Cell Biol.
13
,
159
-
166
.
Tice
,
L. W.
,
Carter
,
R. L.
and
Cahill
,
M. B.
(
1979
).
Changes in tight junctions of rat intestinal crypt cells associated with changes in their mitotic activity
.
Tissue Cell
11
,
293
-
316
.
Trepat
,
X.
,
Deng
,
L.
,
An
,
S. S.
,
Navajas
,
D.
,
Tschumperlin
,
D. J.
,
Gerthoffer
,
W. T.
,
Butler
,
J. P.
and
Fredberg
,
J. J.
(
2007
).
Universal physical responses to stretch in the living cell
.
Nature
447
,
592
-
595
.
Turner
,
J. R.
,
Buschmann
,
M. M.
,
Romero-Calvo
,
I.
,
Sailer
,
A.
and
Shen
,
L.
(
2014
).
The role of molecular remodeling in differential regulation of tight junction permeability
.
Semin. Cell Dev. Biol.
36
,
204
-
212
.
Umeda
,
K.
,
Ikenouchi
,
J.
,
Katahira-Tayama
,
S.
,
Furuse
,
K.
,
Sasaki
,
H.
,
Nakayama
,
M.
,
Matsui
,
T.
,
Tsukita
,
S.
,
Furuse
,
M.
and
Tsukita
,
S.
(
2006
).
ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation
.
Cell
126
,
741
-
754
.
Van Itallie
,
C. M.
,
Holmes
,
J.
,
Bridges
,
A.
,
Gookin
,
J. L.
,
Coccaro
,
M. R.
,
Proctor
,
W.
,
Colegio
,
O. R.
and
Anderson
,
J. M.
(
2008
).
The density of small tight junction pores varies among cell types and is increased by expression of claudin-2
.
J. Cell Sci.
121
,
298
-
305
.
Van Itallie
,
C. M.
,
Fanning
,
A. S.
,
Bridges
,
A.
and
Anderson
,
J. M.
(
2009
).
ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton
.
Mol. Biol. Cell
20
,
3930
-
3940
.
Van Itallie
,
C. M.
,
Tietgens
,
A. J.
,
Krystofiak
,
E.
,
Kachar
,
B.
and
Anderson
,
J. M.
(
2015
).
A complex of ZO-1 and the BAR-domain protein TOCA-1 regulates actin assembly at the tight junction
.
Mol. Biol. Cell
26
,
2769
-
2787
.
Van Itallie
,
C. M.
,
Tietgens
,
A. J.
and
Anderson
,
J. M.
(
2017
).
Visualizing the dynamic coupling of claudin strands to the actin cytoskeleton through ZO-1
.
Mol. Biol. Cell
28
,
524
-
534
.
Van Itallie
,
C. M.
,
Lidman
,
K. F.
,
Tietgens
,
A. J.
and
Anderson
,
J. M.
(
2019
).
Newly synthesized claudins but not occludin are added to the basal side of the tight junction
.
Mol. Biol. Cell
30
,
1406
-
1424
.
Walsh
,
S. V.
,
Hopkins
,
A. M.
,
Chen
,
J.
,
Narumiya
,
S.
,
Parkos
,
C. A.
and
Nusrat
,
A.
(
2001
).
Rho kinase regulates tight junction function and is necessary for tight junction assembly in polarized intestinal epithelia
.
Gastroenterology
121
,
566
-
579
.
Watson
,
C. J.
,
Rowland
,
M.
and
Warhurst
,
G.
(
2001
).
Functional modeling of tight junctions in intestinal cell monolayers using polyethylene glycol oligomers
.
Am. J. Physiol. Cell Physiol.
281
,
C388
-
C397
.
Xu
,
X.
,
Jin
,
D.
,
Durgan
,
J.
and
Hall
,
A.
(
2013
).
LKB1 controls human bronchial epithelial morphogenesis through p114RhoGEF-dependent RhoA activation
.
Mol. Cell. Biol.
33
,
2671
-
2682
.
Yamazaki
,
Y.
,
Tokumasu
,
R.
,
Kimura
,
H.
and
Tsukita
,
S.
(
2011
).
Role of claudin species-specific dynamics in reconstitution and remodeling of the zonula occludens
.
Mol. Biol. Cell
22
,
1495
-
1504
.
Yao
,
M.
,
Qiu
,
W.
,
Liu
,
R.
,
Efremov
,
A. K.
,
Cong
,
P.
,
Seddiki
,
R.
,
Payre
,
M.
,
Lim
,
C. T.
,
Ladoux
,
B.
,
Mege
,
R. M.
, et al. 
(
2014
).
Force-dependent conformational switch of alpha-catenin controls vinculin binding
.
Nat. Commun.
5
,
4525
.
Yonemura
,
S.
(
2011
).
Cadherin-actin interactions at adherens junctions
.
Curr. Opin. Cell Biol.
23
,
515
-
522
.
Yonemura
,
S.
,
Wada
,
Y.
,
Watanabe
,
T.
,
Nagafuchi
,
A.
and
Shibata
,
M.
(
2010
).
alpha-Catenin as a tension transducer that induces adherens junction development
.
Nat. Cell Biol.
12
,
533
-
542
.
Yu
,
A. S.
,
McCarthy
,
K. M.
,
Francis
,
S. A.
,
McCormack
,
J. M.
,
Lai
,
J.
,
Rogers
,
R. A.
,
Lynch
,
R. D.
and
Schneeberger
,
E. E.
(
2005
).
Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells
.
Am. J. Physiol. Cell Physiol.
288
,
C1231
-
C1241
.
Yu
,
D.
,
Marchiando
,
A. M.
,
Weber
,
C. R.
,
Raleigh
,
D. R.
,
Wang
,
Y.
,
Shen
,
L.
and
Turner
,
J. R.
(
2010
).
MLCK-dependent exchange and actin binding region-dependent anchoring of ZO-1 regulate tight junction barrier function
.
Proc. Natl. Acad. Sci. USA
107
,
8237
-
8241
.
Zihni
,
C.
,
Mills
,
C.
,
Matter
,
K.
and
Balda
,
M. S.
(
2016
).
Tight junctions: from simple barriers to multifunctional molecular gates
.
Nat. Rev. Mol. Cell Biol.
17
,
564
-
580
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information