Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


We study punctate adherens junctions (pAJs) to determine how short-lived cadherin clusters and relatively stable actin bundles interact despite differences in dynamics. We show that pAJ-linked bundles consist of two distinct regions-the bundle stalk (AJ-BS) and a tip (AJ-BT) positioned between cadherin clusters and the stalk. The tip differs from the stalk in a number of ways: it is devoid of the actin-bundling protein calponin, and exhibits a much faster F-actin turnover rate. While F-actin in the stalk displays centripetal movement, the F-actin in the tip is immobile. The F-actin turnover in both the tip and stalk is dependent on cadherin cluster stability, which in turn is regulated by F-actin. The close bidirectional coupling between the stability of cadherin and associated F-actin shows how pAJs, and perhaps other AJs, allow cells to sense and coordinate the dynamics of the actin cytoskeleton in neighboring cells-a mechanism we term "dynasensing."

Free full text 


Logo of nihpaLink to Publisher's site
Cell Rep. Author manuscript; available in PMC 2020 Mar 24.
Published in final edited form as:
PMCID: PMC7093076
NIHMSID: NIHMS1566857
PMID: 32101754

Sensing Actin Dynamics through Adherens Junctions

SUMMARY

We study punctate adherens junctions (pAJs) to determine how short-lived cadherin clusters and relatively stable actin bundles interact despite differences in dynamics. We show that pAJ-linked bundles consist of two distinct regions—the bundle stalk (AJ-BS) and a tip (AJ-BT) positioned between cadherin clusters and the stalk. The tip differs from the stalk in a number of ways: it is devoid of the actin-bundling protein calponin, and exhibits a much faster F-actin turnover rate. While F-actin in the stalk displays centripetal movement, the F-actin in the tip is immobile. The F-actin turnover in both the tip and stalk is dependent on cadherin cluster stability, which in turn is regulated by F-actin. The close bidirectional coupling between the stability of cadherin and associated F-actin shows how pAJs, and perhaps other AJs, allow cells to sense and coordinate the dynamics of the actin cytoskeleton in neighboring cells—a mechanism we term “dynasensing.”

In Brief

Indra et al. show that actin bundles at adherens junctions consist of a stalk and a cadherin-interacting tip. F-actin is relatively stable in the stalk, but turns over rapidly in the tip, and turnover rates in both depend on associated cadherin clusters. This bidirectional coupling may allow neighboring cells to coordinate their actin dynamics.

INTRODUCTION

Cadherin-mediated adhesion is a defining trait of all metazoans. It is mediated by a transmembrane receptor, a classical cadherin (E-cadherin in many epithelia), which functions in a large multi-protein complex. The unstructured cytoplasmic region of E-cadherin binds directly to β-catenin, which in turn is bound to α-catenin (we denote this as the cadherin–catenin complex, or CCC). The CCC-incorporated α-catenin also interacts with F-actin through its actin-binding domain (Pappas and Rimm, 2006; Harris and Tepass, 2010; Michael and Yap, 2013; Mège and Ishiyama, 2017), providing a bridge between cadherin and the actin cytoskeleton. We recently showed that the extracellular region of punctate adherens junctions (pAJs) consists of dense, paracrystalline nanoclusters formed through cis and trans interactions of cadherin ectodomains, interspersed with less dense cadherin regions. The nanoclusters in this “mosaic model” assemble and disassemble on a time-scale of seconds in a process that depends on the dynamics of junctional actin (Indra et al., 2018). However, the pAJs are linked to F-actin bundles, which are thought to be among the most stable actin structures. How then can the fast dynamics of cadherin clusters and the much slower turnover of actin bundles be reconciled? Further, the pulling forces the bundles apply on the CCC are essential for both the overall stability of AJs and the tension-dependent recruitment of vinculin and other proteins into AJs (Huveneers and de Rooij, 2013; Mège and Ishiyama, 2017). How can these tensile forces be applied between regions with very different dynamical properties? Here we address these questions by studying the spatiotemporal properties of E-cadherin clusters and F-actin bundles in pAJs.

AJs, including pAJs, along with focal adhesions (FAs) and tips of lamellipodia, are sites of rapid F-actin polymerization (Vasioukhin et al., 2000; Zhang et al., 2005; Kovacs et al., 2011). However, the polymerization mechanism and the role and fate of polymerized filaments in AJs are poorly understood. In one model, F-actin at AJs is nucleated by the Arp2/3 complex. Direct binding of Arp2/3, to the cytoplasmic tail of E-cadherin (Kovacs et al., 2002), or indirect binding through cortactin (Han et al., 2014), neogenin (Lee et al., 2016), or α-actinin-4 (Tang and Brieher, 2012) have all been suggested as mechanisms for Arp2/3 recruitment into AJs. The Arp2/3-nucleated filaments have been proposed to grow using Ena/Vasodilator-stimulated phosphoprotein(VASP) proteins (Scott et al., 2006; Leerberg et al., 2014) and to then be reconfigured from a branched to a bundled array by an N-WASP-dependent mechanism (Kovacs et al., 2011; Brieher and Yap, 2013). This model is challenged, however, by observations that mature AJs are depleted of Arp2/3 (Verma et al., 2004; Hansen et al., 2013) and by proteomics analyses that have failed to identify Arp2/3, neogenin, N-WASP, or α-actinin-4 in association with cadherin tails (Van Itallie et al., 2014; Guo et al., 2014; Li et al., 2019).

There are also uncertainties concerning the configuration and dynamics of F-actin in AJs. Platinum replica electron microscopy (EM) identified a pool of branched F-actin at different types of AJs, including at pAJs, of endothelial cells (Efimova and Svitkina, 2018), while other EM studies have suggested that AJs are directly associated with tensile actin bundles (Yonemura et al., 1995; Buckley et al., 2014; Choi et al., 2016; Li et al., 2019). With respect to dynamics, fluorescence recovery after photobleaching (FRAP) and fluorescence loss after photoactivation (FLAP) experiments with Madin Darby Canine Kidney (MDCK) cells showed that AJ-associated F-actin recovers with a half-life of seconds (Yamada et al., 2005), a rate that corresponds to that of the branched F-actin in lamellipodia (Lai et al., 2008; Fritzsche et al., 2013). However, other studies have reported a half-life of ~1 min or even longer (Wu, 2014; Cavey et al., 2008; Kovacs et al., 2011), corresponding to that of bundled rather than branched actin (Hotulainen and Lappalainen, 2006).

Here we show that pAJ-associated bundles are composed of two distinct regions: a stable F-actin bundle, which we refer to as the adherens junction bundle stalk (AJ-BS), and a highly dynamic pAJ-proximal region at the tip of this bundle, the adherens junction bundle tip (AJ-BT). The existence of these two connected but distinct actin bundle regions helps to reconcile previous studies, which have alternatively found both branched and dynamic, and bundled and stable, F-actin in AJs. Since the dynamical properties of the AJ-BTs are similar to those of CCC clusters, the existence of the tips resolves the issue of a mismatch between bundle and CCC dynamics. Finally, bidirectional coupling between CCC and F-actin dynamics coordinates organization of the actin cytoskeleton in adjacent cells. Our work, therefore, provides a dynamical model of pAJs, which explains how they are able to maintain and synchronize their remarkable flexibility under constant actomyosin stretching. In light of their common protein compositions, this model may be applicable to AJs in general.

RESULTS

AJ-linked Actin Bundles Consist of Two Distinct Regions with Differing Compositions of Actin-Binding Proteins

The pAJs are the most prominent AJs of human squamous carcinoma A431 cells (Indra et al., 2013, 2018), and are identified by E-cadherin staining in Figure 1A. Phalloidin staining showed that they are linked to radial actin bundles of diverse length, which approach each pAJ from two adjacent cells. Their pAJ-attached termini exhibit the strongest phalloidin fluorescence (Figure 1A, arrowheads), perhaps due to overlap of the actin bundles from each cell linked at the junction. The extra-junctional end of the bundle appears to integrate into a dense actin cortical network (Figure 1A).

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0002.jpg
Organization of pAJ-linked Actin Bundles in A431 Cells

(A–D) Projections of all x-y optical slices of A431 cells stained for (A) E-cadherin (Ecad, red) and F-actin (actin, green); (B) Ecad (green), F-actin (red), and myosin IIA (myosin II, blue); (C) Ecad (green), actin, and calponin-3 (Cnn3); and (D) Cnn3 (green) and myosin IIA (red).

Scale bar, 10 μm. The zoomed areas (indicated by dashed boxes) are presented on right panel. Scale bar, 5 μm. Group of pAJs in a left panel in (A) is further magnified in the inset. Arrowheads in (A)–(C) indicate adherens junction bundle tips (AJ-BTs), which are enriched with F-actin and Ecad, but are devoid of myosin IIA and Cnn3. Arrowhead in (D) shows that myosin IIA is not integrated into a Cnn3-positive pAJ-linked bundle.

To better understand the organization of the pAJ-connected bundles, we stained A431 cells for myosin IIA (MyoIIA) and calponin-2 or −3 (Cnn2/3). These proteins have been shown to be abundant in actin bundles in fibroblasts (Tojkander et al., 2012; Ciuba et al., 2018). We found that MyoIIA was typically concentrated several microns away from pAJs (Figure 1B). By contrast, Cnn2/3 were specifically enriched in the pAJ-connected bundles (Figures 1C and S1). However, their pAJ-integrated termini were completely devoid of Cnn2/3 (Figure 1C, arrowhead). Double Cnn3/MyoIIA staining confirmed that calponin-positive pAJ-linked bundles did not incorporate MyoIIA (Figure 1D).

The absence of Cnn2/3 staining in the pAJ-attached termini suggests that these regions are structurally different from the remaining parts of the bundles; however, this observation could also be due to inaccessibility of Cnn2/3 to antibodies at sites of actin-CCC interactions. To distinguish these two possibilities, we expressed recombinant Dendra2-tagged Cnn3 (Cnn3-Dn) in A431 cells. Figures 2A and and2B2B show that, consistent with the antibody staining, Cnn3-Dn was efficiently incorporated into the pAJ-linked bundles, but not into their pAJ-integrated termini. Line-scan analysis of fluorescence intensity across the pAJs confirmed that the depressions in Cnn3-Dn incorporation exactly matched the peaks of both F-actin and E-cadherin (Figure 2E).

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0003.jpg
F-actin in AJ-BT Regions of pAJ-linked Bundles Associates with a Distinct Set of Actin-Binding Proteins

(A–D) Projections of all x-y optical slices of Cnn3-Dn-expressing A431 cells imaged for Dendra2 fluorescence (green, Cnn3-Dn) and stained for (A) Ecad, (B) actin, (C) vinculin (Vcn), and (D) mena. Scale bar, 10 μm. The zoomed areas (indicated by dashed boxes in Cnn3-Dn images) are presented at the bottom. Scale bar, 5 μm. Arrowheads indicate the gaps in Cnn3-Dn distribution along pAJ-linked bundles that are precisely matched by the antibody staining.

(E) Line-scan analysis of the selected pAJs for distribution of Cnn3-Dn (green) and the indicated pAJ-associated proteins revealed by specific antibodies (red).

(F) Structured Illumination Microscopy of Cnn3-Dn-expressing A431 cells double-stained for Dendra2 (green, Cnn3) and Ecad (red). Arrowhead indicates AJ-BT region. Scale bar, 10 μm. The zoomed areas (indicated by dashed boxes in Cnn3-Dn images) are presented at the bottom. Scale bar, 5 μm.

(G) The thickness of the Cnn3-Dn-deficient regions measured for 15 independent pAJs on Ono, 2018; Wu, 2014 SIM and on confocal images (mean +/−SEM). Two-tailed t test.

(H) Schematic representation of pAJ-linked bundles. Each bundle appears to consist of at least two regions. The first region, AJ-BT (AJ-BT), which is Cnn2/3-deficient (CNN2/3-), directly contacts the cadherin-catenin complex (CCC). The second, Cnn2/3-positive (CNN2/3+) region, AJ-BS, is attached to AJ-BT and interconnects the AJs with the actomyosin cytoskeleton.

We next tested whether major actin cytoskeletal proteins associated with pAJs—vinculin, VASP, mena, and FBLIM1—were located at the Cnn3-Dn–deficient regions of the pAJ-linked bundles. Indeed, immunofluorescence microscopy of Cnn3-Dn–expressing cells for these proteins, and line-scan analyses of the resulting images, clearly showed the presence of these proteins in the pAJ-adjacent bundle termini (Figures 2C2E and S1). Structured-illumination microscopy (SIM) of E-cadherin, which marks the locations of pAJs, further validated localization of E-cadherin within the Cnn3-Dn–deficient regions of the bundles (Figure 2F). The average thickness of this region, assessed using 30 independent pAJ images (obtained by SIM or conventional microscopy), was ~400 nm (Figure 2G).

Overall, these results show that a pAJ-linked actin bundle is structurally heterogeneous and encompasses at least two regions (Figure 2H). The first region overlaps with the CCC at the light-microscopy resolution level. It exhibits highest F-actin deposition, is enriched with VASP, mena, FBLIM1, and vinculin, and is devoid of Cnn2/3. We denote this region as the bundle tip (AJ-BT), which, given the 400-nm total length of the Cnn2/3-deficient area, has a length in each cell on the order of 200 nm. The second region corresponds to the bundle stalk (AJ-BS), which is enriched with Cnn2/3. The distal end of AJ-BSs interacts with the AJ-BT while the opposite end is integrated into the contractile actomyosin network, which ultimately transmits the mechanical forces into this system.

F-actins in AJ-BSs and AJ-BTs Have Different Dynamical Properties

The relatively large size of the tip region allows it to be analyzed using conventional light microscopy. To this end, we produced A431 cells that stably express a photoactivatable form of actin, mCHPA-actin. Previous studies have shown that actin tagged with fluorescent proteins assembles into filaments and can serve as an excellent probe of F-actin dynamics (Yamada et al., 2005; Tojkander et al., 2015). Indeed, double immunostaining of the transfected cells for mCHPA-actin and phalloidin showed that the actin bundles of these cells recruited the recombinant actin (Figure 3A). Furthermore, similar to the untagged actin in the control cells, mCHPA-actin produced bright plaques at sites of E-cadherin-based pAJs (Figure 3B). To specifically photoactivate mCHPA fluorescence at bundle tips or at bundle stalks, we developed a “pAJ-actin dynamics” assay in which the mCHPA-actin cells were co-cultured with mGFP-tagged E-cadherin (EcGFP)-expressing A431 counterparts (see Figure 3C). These mCHPA-actin/EcGFP co-cultures allowed us to study actin dynamics in mixed pAJs, one half of which was highlighted by EcGFP, with the other half expressing the “dark” mCHPA-actin.

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0004.jpg
Dynamics of F-actin in AJ-BTs and in AJ-BSs

(A and B) Maximum projection view of A431 cells stably expressing mCHPA-actin stained for (A) actin using Phalloidin (actin, green) and mCHPA-actin using anti-mCH antibody (mCH, red) and (B) Ecad (green) and mCHPA-actin (red). Scale bar (in A and B), 10 μm. The zoomed areas (indicated by dashed boxes in actin and Ecad images) are presented at the bottom. Scale bar, 5 μm.

(C) Time-lapse series of mCHPA-actin photoactivated in the co-cultures of mCHPA-actin- and EcGFP-expressing cells. The series were taken in green and red imaging channels: green channel detects EcGFP, while red channel detects only background fluorescence before photoactivation. The full-screen images (left) show groups of cells just before photoactivation. Scale bar, 10 μm. The time-lapse frames of the boxed areas are zoomed at the right panels and the arrowheads show the selected pAJs before photoactivation (−1), immediately after (0) and then at particular following time points (exact time is indicated in s). Red fluorescence of mCHPA-actin was activated in diffraction-limited spots (~500 nm in diameter) in three different locations: exactly at sites of EcGFP-tagged pAJs (AJ-BT); ~0.5 μk toward the center of the dark cell (AJ-BS), or at the pAJ lateral sides (lateral to AJ). Scale bar, 5 μm.

(D) Kinetics of decay of mCHPA-actin photoactivated in AJ-BT, AJ-BS, and lateral to pAJ (n = 10, mean +/− SEM).

(E) Parameters for mCHPA-actin fluorescence decay assessed from curves presented in (D).

(F) Displacement of individual photoactivated spots of actin over 1 min (n = 10). Two-tailed t test.

First, we photoactivated mCHPA precisely at pAJ sites using the maximum focused laser beam (Figure 3C, AJ-BT; Video S1). Time-lapse imaging showed that at this position, the spot of red mCHPA-derived fluorescence (its diameter encompassed ~4 to 5 pixels, or 440–550 nm) exhibited a relatively short lifetime, with the half-life of fluorescence decay ~20 s, with low variation between experiments (Figures 3D and and3E).3E). The photoactivated spot, usually trackable for up to 1 min, was immobile and tightly associated with the selected pAJ during the entire observation period (Figure 3F). To probe actin dynamics in the AJ-BSs, we targeted the laser beam to sites located ~0.5 μm from the pAJ toward the center of the dark cell (Figure 3C, AJ-BS; Video S2). Remarkably, at these sites the decay of photoactivated spots was significantly slower and furthermore, in most cases the spots showed retrograde movements (Figure 3F). Both of these features, the retrograde movement (median ~400 nm/min) and the rate of fluorescent decay (median t1/2 ~50 s) significantly varied between individual junctions (Figures 3E and and3F).3F). To ensure that the F-actin dynamic features were specific for the selected spots along pAJ-linked bundles, we performed control experiments, which targeted sites near to, but outside of, the pAJs. At these locations, where junctional actin is not present (Figures 3C and and3D;3D; Video S3), the half-life of the resulting red fluorescence decay (~10 s) was significantly faster than in AJ-BTs.

Taken together, the pAJ-actin dynamics assay confirmed the compound architecture of the pAJ-linked actin bundles. Their AJ-BTs appear to consist of short-lived F-actin whose turnover rate of ~20 s is similar to that of lamellipodium actin (Fritzsche et al., 2013; Lai et al., 2008). By contrast, F-actin in AJ-BSs exhibited characteristics more similar to those in dorsal actin bundles linked to FAs, which display centripetal flow with an average half-life of ~2 min (Hotulainen and Lappalainen, 2006; Tojkander et al., 2015). While we cannot exclude the possibility that a small pool of AJ-BT-linked F-actin eventually moves into the AJ-BS, the bulk of the AJ-BT F-actin was immobile and tightly coupled with the CCC in pAJs.

Cadherin Cluster Stability Regulates F-actin Turnover in AJ-BTs and AJ-BSs

The fast turnover of AJ-BT F-actin suggests that the assembly-disassembly of both F-actin and cadherin clusters is tightly coupled. This idea is supported by previous observations that stabilization of F-actin by Jasplakinolide prolonged the lifetime of cadherin clusters in pAJs (Indra et al., 2018). Does the turnover of F-actin in AJ-BTs depend on the stability of cadherin clusters? To answer this question, we studied how different manipulations known to affect cadherin cluster dynamics influence that of F-actin. The green fluorescence in one half of pAJs in our mCHPA-actin/EcGFP co-cultures allowed us to use FRAP technique to determine the dynamical properties of cadherin in pAJs. While FRAP significantly underestimates the rate of cadherin cluster assembly and disassembly due to local reincorporation of cadherin into new clusters (Indra et al., 2018), this approach is useful for the general assessment of cadherin cluster stability. First, we expose the co-cultures to hypertonic shock, which we have shown to dramatically reduce cadherin dynamics in pAJs (Troyanovsky et al., 2006). Indeed, after 10 min in hypertonic sucrose media, the recovery of EcGFP fluorescence in pAJs as well as the decay of mCHPA-actin photoactivated fluorescence in AJ-BTs was nearly completely stalled (Figures 4A and and4B).4B). Interestingly, the mCHPA-actin turnover in AJ-BSs as well as its retrograde flow were also blocked (Figures 4B and and4C4C).

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0005.jpg
Cadherin Regulates F-actin Turnover in pAJ-linked Bundles

(A) FRAP of EcGFP in pAJs after 10 min in hypertonic sucrose media (mean +/− SEM).

(B) Decay of mCHPA-actin photoactivated in AJ-BT (actin-AJ-BT) or in AJ-BS (actin-AJ-BS) (mean +/− SEM).

(C) Displacements of the photoactivated spot as in Figure 3F.

(D) Maximum projection view of three different co-cultures of A431 cells. One of the sublines stably expresses mCHPA-actin, which is visualized by anti-mCH staining (mCH, red). These cells form mixed pAJs with cells stably expressing EcGFP (GFP, green), or its mutants, cis mutant EcGFP (cis-GFP) and P5A-EcGFP (P5A-GFP). Scale bar, 10 μm. The zoomed areas (indicated by dashed boxes in green images) are presented on the right panel. Scale bar, 5 μm.

(E) FRAP of pAJs in cells expressing EcGFP or its mutants P5A-EcGFP or cis mutant EcGFP (n = 15). FRAP was performed either in homogeneous cultures of corresponding cells (Homogeneous pAJ) or in co-cultures with “dark” mCHPA-actin-expressing A431 cells. In the latter case the pAJs located between two types of cells were studies (mean +/− SEM).

(F) Decay of mCHPA-actin spots photoactivated in pAJs (Actin-AJ-BT) or in bundles (Actin-AJ-BS) in co-cultures with EcGFP or its mutants. Right panel shows parameters of corresponding decays (mean +/− SEM).

(G) Displacements of the photoactivated spots as in (C).

Since hypertonic shock abolished pAJ disassembly through relatively indirect and still unknown mechanisms, we chose to modify cadherin cluster stability directly using specific point mutations that effect cadherin ectodomain trans and cis interactions. Specifically, in the mCHPA-actin/EcGFP co-culture, we replaced the EcGFP cells with cells expressing mutant EcGFP. The first, involving the P5A mutant, dramatically strengthens the KD of cadherin trans-bonds, from ~100 μM to 3 μM (Vendome et al., 2011), and thus increases cluster stability. Notably, since P5A forms “extra” hydrogen bonds in trans with distal β-strand residues, the strengthening effect of this mutation should also be observed when binding to wild-type (WT) E-cadherin (Vendome et al., 2011). The second mutant targets the cis-binding interface of E-cadherin and destabilizes pAJs by disrupting the paracrystalline lattice formed by cadherin ectodomains (Harrison et al., 2011; Strale et al., 2015).

We first performed a set of control experiments in order to confirm that in the mixed pAJs in our co-cultures, the P5A-EcGFP mutant would indeed decrease, and the EcGFP cis mutant would increase, the turnover rate of the intact E-cadherin present in the mCHPA-actin-expressing cells. We showed that both mutants established mixed pAJs with mCHPA-actin-expressing cells and that these mixed pAJs were linked to the mCHPA-actin bundles (Figure 4D). Then, using a FRAP assay with the mutant-expressing cells in homogeneous cultures and in co-cultures with mCHPA-actin-expressing cells, we verified that both mutants changed EcGFP turnover rates as expected. Indeed, fluorescence recovery curves (Figure 4E) showed that the P5A mutation significantly reduced, while the cis mutation, by contrast, increased, the pAJ cadherin turnover rate in both the mixed and homogeneous pAJs. The P5A mutant likely stabilizes mixed pAJs through enhanced trans interactions with WT E-cadherin, whereas the cis mutant destabilizes pAJs likely by disrupting one half of the E-cadherin lattice, thus disrupting the other half.

Finally, we confirmed that turnover of the WT E-cadherin on one half of pAJs matched that of the mutant E-cadherin present on the opposite side of the same pAJ. To show this, we co-cultured the mutant-expressing cells with A431 cells expressing mCH-tagged E-cadherin (EcmCH). Both, GFP and mCH fluorophores in the hetero-chromatic pAJs were simultaneously bleached and recovery of their fluorescence simultaneously monitored by time-lapse microscopy (Figure S2). This two-color FRAP experiment revealed that E-cadherin turnover rates were indeed balanced in both sides of AJs, consistent with previously published data showing that cadherin turnover rates are closely correlated in two halves of the same junction (de Beco et al., 2015).

These observations confirmed that the replacement of EcGFP with P5A-EcGFP or with cis-EcGFP, respectively, either slowed or increased WT E-cadherin turnover in the mCHPA-actin-expressing cells connected through mixed pAJs in our co-cultures. Next, using our pAJ-actin dynamics assay, we determined actin turnover rates in these junctions. The results clearly showed that AJ-BTs from the pAJs for which the opposite half incorporated the P5A-EcGFP mutant exhibited a significant increase in the lifetime of mCHPA red fluorescence (t1/2 ~40 s). By contrast, mCHPA fluorescence in AJ-BTs for which pAJs contacted cis-EcGFP-expressing cells had much shorter lifetimes, with t1/2 of just few seconds (Figure 4F). Surprisingly, both mutations correspondingly changed the F-actin turnover not only in the AJ-BTs, but also in the AJ-BSs (Figure 4F). Finally, the centripetal flow of the red spot photoactivated at the AJ-BSs was nearly completely stalled in the cells connected to P5A-EcGFP but, in contrast, increased in cis mutant EcGFP co-cultures (Figure 4G).

F-actin Dynamics Are Coordinated between Adjacent Cells

Our previously reported data suggest that F-actin turnover regulates the rate of cadherin cluster assembly and disassembly (Indra et al., 2018). Here we have demonstrated that the inverse phenomenon also takes place: cadherin cluster dynamics control the rate of F-actin turnover in pAJ-linked bundles. These observations imply that F-actin polymerization–depolymerization rates in both halves of a pAJ are coordinated. To test this hypothesis we assessed whether, in the mCHPA-actin/EcGFP co-culture, F-actin stabilization in the EcGFP expressing cells would stabilize F-actin in the adjacent mCHPA-actin-expressing cells.

To stabilize F-actin in the EcGFP-expressing cells, we knocked out a key actin depolymerizing factor, cofilin 1 (Cfl1), using a CRISPR/Cas9 approach. The lack of Cfl1 has been shown to reduce the F-actin depolymerization rate of the branched actin network of different types of cells, but does not abolish AJs (Lappalainen and Drubin, 1997; Hotulainen et al., 2005; Iwasa and Mullins, 2007; Vitriol et al., 2013; Wang et al., 2016). As has been shown for other cell types, Cfl1 knockout (KO) in the EcGFP-expressing A431 cells resulted in cell shape flattening, which may be caused by unbalanced actin polymerization in cell lamellipodia. Despite these changes, as has been observed previously (Chu et al., 2012; Wang et al., 2016), the Cfl1-KO cells retained their pAJs (Figure 5A) and general actin bundle organization, as revealed by phalloidin and anti-Cnn3 staining (Figure 5B, ,C).C). As expected from our previous experiments with Jasplakinolide (Indra et al., 2018), the FRAP assay showed a significant stabilization of E-cadherin in AJs upon Cfl1 KO (Figure 5D, homogeneous pAJ).

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0006.jpg
Knockout of Cfl1 Affects Actin Dynamics in the Neighboring WT Cells

(A–C) Maximum projection view of control A431-EcGFP cells (A431-EcGFP) or after Cfl1-KO (A431-EcGFP-Cfl1-KO). Cells were imaged for GFP (EcGFP) and for (A) Cfl1 (Cfl, red), (B) actin (actin, red), and (C) Cnn-3 (Cnn3, red). Scale bar, 10 μm. The zoomed areas (indicated by dashed boxes in EcGFP images) are presented in the insets (A) or at the bottom (B and C). Scale bar, 5 μm.

(D) FRAP of pAJs in control and in Cfl1-KO A431-EcGFP cells (n = 15). FRAP was performed either in homogeneous cultures of corresponding cells (homogeneous pAJ) or in co-cultures with “dark” mCHPA-expressing A431 cells. In the latter case the pAJ between dark cells and cells expressing EcGFP were studied. The corresponding FRAP parameters are shown at the bottom (mean +/− SEM).

(E) Decay of mCHPA-actin spots photoactivated in pAJs (actin-AJ-BT) or in bundle stalks (actin-AJ-BS) in mCHPA-actin expressing cells co-cultured with A431-EcGFP or A431-EcGFP-Cfl1-KO cells. The corresponding parameters of mCHPA-actin turnover are shown at the bottom (mean +/− SEM).

EcGFP in the Cfl1-KO cells showed nearly the same turnover rate in the co-culture with mCHPA-actin-expressing cells as in the homogeneous culture (Figure 5D). As expected from this observation, photoactivation of mCHPA-actin in such co-cultures showed that the Cfl1-KO EcGFP-expressing cells significantly stabilized actin in both AJ-BTs and AJ-BSs in adjacent cells (Figure 5D). Together, these experiments suggest that the reduction of F-actin dynamics induced by Cfl1-KO in EcGFP-expressing cells was transduced into the neighboring cells through reduction of cadherin turnover in AJs.

pAJs Are Enriched with Factors Driving Dynamics of the Branched Actin Cytoskeleton

To further shed light on the organization of pAJ-associated actin filaments and mechanisms of their fast dynamics, we stained A431-EcGFP cells for proteins known to bind and regulate the dynamics of branched actin filaments. First, we extended our examination of Cfl1. This protein transiently targets and severs actin filaments in collaboration with another protein, Aip1 (Bravo-Cordero et al., 2013; Ono, 2018). While, as observed for other cell types (Abe et al., 1993; Munsie et al., 2012), Cfl1 was mostly cytosolic in A431 cells, many of the well-isolated pAJs exhibited Cfl1-specific puncta (Figure 6A). Line-scan analyses confirmed close spatial association between pAJs and such Cfl1 puncta (Figures 6A and and6E).6E). It has been shown that in absence of Aip1, Cfl1 binds to but does not sever filaments, leading to accumulation of Cfl1-bound F-actin (Kato et al., 2008); we exploited this observation to clearly identify the pool of Cfl1-targetted actin filaments in Aip1 knockout A431-EcGFP cells. As shown in Figure 6B, these cells exhibited numerous Cfl1-decorated cortical filaments, which were especially prominent in pAJs (Figure 6B). The strong enrichment of Cfl1 in pAJ-linked filaments, together with the dramatic reduction of actin and E-cadherin turnover in pAJs upon Cfl1 KO, strongly suggests that Cfl1 plays a direct and critical role in F-actin homeostasis in pAJs.

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0007.jpg
Subcellular Localization of Proteins Involved in Branched Actin Dynamics: Cfl1, Arp2/3, and Cortactin

Projections of all x-y optical slices of A431-EcGFP cells

(A), (C), (D), and A431-EcGFP-Aip1-KO cells (B) imaged for GFP fluorescence (green, EcGFP) and stained for Cfl1 (Cfl), Aip1 (Aip), Arp2/3 (Arp2/3), and cortactin (CTTN). Scale bar, 10 μm. The zoomed areas (indicated by dashed boxes in EcGFP images) are presented at the bottom or in the insets (for anti-Aip staining in A). Arrowheads indicate pAJs, which exhibit clear colocalization with the antibody-stained proteins. Arrows marked pAJs, which colocalization with the stained protein is unclear because of high nearby fluorescence. Scale bar, 5 μm.

(E) Line-scan analysis of the pAJs (selected by arrowheads in A–D) for distribution of EcGFP (green) and the indicated pAJ-associated proteins (red). In the case of Arp2/3 and cortactin, two line-scans are shown—one for “marked by arrowhead” and another for “marked by arrow” as pAJs.

Next we studied localization of Arp2/3, a key protein complex involved in organization and dynamics of the branched actin cytoskeleton. As highlighted in the Introduction, there are strong uncertainties about the mechanisms of Arp2/3 recruitment into AJs and even about its presence in mature AJs (Verma et al., 2004; Hansen et al., 2013). This uncertainty stems in part from the fact that Arp2/3 is abundant in the cell cortex and is especially enriched in cell–cell contact areas. To reduce cytosolic pools of Arp2/3, we fixed cells in cytoskeleton-preserving buffer in the presence of Triton X-100. Figures 6C and and6D6D show that Arp2/3 and its cofactor, cortactin, were highly concentrated in the cell–cell contact regions. In contrast to Cfl1, for which peak intensity usually corresponded to that of E-cadherin, Arp2/3 and cortactin were located in relatively large zones encompassing the pAJs (Figure 6D). These results suggest that Arp2/3, in agreement with proteomics data, might not be specifically targeted into mature pAJs. Instead, Arp2/3 might be involved in formation and dynamics of the entire actin cortex of the lateral membranes in which the AJ-BTs are integrated. More work is needed to understand whether Arp2/3 plays a specific role in generation and dynamics of CCC-coupled F-actin.

DISCUSSION

In this work we show that pAJ-linked actin bundles consist of two distinct regions: a bundle stalk, the AJ-BS, which connects the bundle to the actomyosin cytoskeleton; and a bundle tip, the AJ-BT, which serves as a joint between the relatively stable stalk and the highly dynamic adhesive region of pAJs. Our FLAP experiments show that the half-life of F-actin in AJ-BTs is ~20 s as compared to ~1 min in AJ-BSs. Time-lapse recordings of these adjacent F-actin regions suggest that they are generated by distinct F-actin assembly pathways since mCHPA-actin activated in AJ-BTs does not translocate into the AJ-BS, but remains associated with corresponding pAJs over its entire lifetime. The F-actin in the AJ-BS, by contrast, is not immobile but flows rearward.

These differences in dynamics may reflect the striking structural difference between these two bundle compartments. The AJ-BS apparently consists of parallel actin filaments since it is enriched with the prominent actin bundling proteins Cnn2/3. The AJ-BT lacks these bundling proteins, but instead is enriched with proteins involved in actin dynamics, such as mena and VASP. The F-actin depolymerization factor, Cfl1, which plays a key role in the dynamics of the branched actin networks (Chan et al., 2009; Bravo-Cordero et al., 2013), is also colocalized with AJ-BTs. The role of this factor in AJ-BT dynamics is supported by the dramatic stabilization of these structures upon Cfl1 KO. AJ-BTs are integrated into the cell–cell contact areas enriched with Arp2/3, known to play a role in nucleation of branched actin networks. This dual organization of pAJ-linked actin bundles is consistent with a recent platinum replica EM study of endothelial AJs, which shows that bundles of parallel actin filaments are linked to a VE-cadherin-rich zone through a ~200–300-nm-thick branched actin network (Efimova and Svitkina, 2018).

Our study also reveals that during its entire lifespan F-actin photoactivated in AJ-BTs associates with pAJs. This observation is important since it suggests that assembly-disassembly cycles of F-actin in AJ-BTs are coupled with those of cadherin clusters to which they are linked via α-catenin. This idea is further reinforced by the observed bidirectional relationship between F-actin and cadherin turnover. In all cases when F-actin was stabilized, E-cadherin clusters also showed increased stability. Our study provides two examples of this relationship: KO of the F-actin depolymerization factor Cfl1 and osmotic shock all simultaneously increased the lifetime and immobile fraction of both E-cadherin and F-actin in AJ-BTs. These observations are in line with previous data showing that F-actin stabilization by Jasplakinolide increases stability of E-cadherin clusters in AJs (Indra et al., 2018). Here we have shown that the inverse is also true: increases or decreases in the lifetime of E-cadherin clusters in pAJs induced by mutant E-cadherins correspondingly changed F-actin lifetimes.

The precise spatiotemporal organization of F-actin in AJ-BTs and the mechanisms responsible for the close coupling between actin and cadherin cluster turnover are still unclear and are important avenues for future research. Figure 7 presents a very general schematic model, based on the results published here and previously (Indra et al., 2018; Efimova and Svitkina, 2018), as to how F-actin and cadherin cluster dynamics are coupled and coordinated. The figure illustrates how parallel and stable actin filaments of the AJ-BS interact with apparently branched and dynamic actin filaments in the AJ-BT, which in turn, interact with the CCC. Our results strongly suggest that each cadherin nanocluster associates with its own “tangle” of the branched F-actin network. The top panel depicts two cadherin nanoclusters linked to two such tangles that are separated by a region of lower cadherin and actin density. In the bottom panel, these two tangles and associated CCC clusters depolymerize and a new tangle linked to a new cadherin cluster is formed. The transition between the structures shown in the two panels involves the rearrangement of cadherin clusters that continuously dissociate and reform in pAJs as a result of two coupled processes that are each cooperative: an extracellular process involving cis and trans cadherin interactions (Harrison et al., 2011; Strale et al., 2015) and an intracellular process involving α-catenin binding to F-actin (Hansen et al., 2013; Buckley et al., 2014; Chen et al., 2015). It is possible that Arp2/3 and Cfl1 play key roles in this process. Importantly, new cadherin clusters continuously reutilize E-cadherin of the disassembled clusters, which results in underestimation of cadherin turnover rate by FRAP or FLAP assays (Indra et al., 2018). By contrast, the requirement for ADP–ATP exchange impedes the immediate reuse of the G-actin released from the disassembled F-actin tangles. This difference in assembly mechanisms could be a major reason for significant differences in the half lives of E-cadherin and AJ-BT actin in pAJs (~5 min and ~20 s, respectively).

An external file that holds a picture, illustration, etc.
Object name is nihms-1566857-f0008.jpg
Schematic Model of Coupling between F-actin and Cadherin in pAJs

A hypothetical cross section of a pAJ is shown. Blue parallel lines represent the actin bundle stalk (AJ-BS) apparently consisting of parallel actin filaments of the same polarity. The stalk is terminated with the bundle tip (AJ-BT) shown by red intersecting lines, which in turn interact with the CCCs (green). Initially (the top image) the bundle tip consists of two cadherin clusters (1 and 2), formed in association with corresponding tangles of branched F-actin. In the bottom panel, these tangles and associated clusters are disassembled and a new CCC cluster (3) and a corresponding new actin tangle are assembled. The new cluster recruits only a few new cadherin molecules (yellow), but the new tangle is built entirely from newly delivered G-actin (magenta). The AJ-BT/AJ-BS interface is positioned to play a key role in dynamics and/or organization of both AJ-BT and AJ-BS.

Close coupling between cadherin clusters and F-actin in AJ-BTs resolves the question highlighted in the Introduction about the apparent mismatch between cadherin and F-actin turnovers in pAJs. Furthermore, it explains how tensile actomyosin-based forces conducted by the AJ-BS can be applied to the “fluid” cadherin-based adhesive interface of pAJs: At each given moment, the interface contains many cadherin-adhesive clusters that are each linked to the tensile AJ-BS through their own tangles of F-actin. A new question now arises, however, as our findings show that there is indeed a mismatch, but it is between AJ-BT and AJ-BS F-actin dynamics. This mismatch suggests that the AJ-BS/AJ-BT interface has novel properties, which are yet to be fully understood. We speculate that the AJ-BS/AJ-BT interface may correspond to a two-sided platform, which continuously produces short-lived tangles of branched F-actin on its AJ-BT side and stable parallel filaments on the opposite, AJ-BS side. Indeed, the centripetal flow we observed for photoactivated actin in AJ-BS suggests active actin polymerization within the bundle, including at the AJ-BS/AJ-BT interface. This polymerization, together with pulling forces generated by MyoIIA, could underlie the rearward treadmilling along bundles we observed for photoactivated AJ-BS actin spots. Interestingly, both the kinetics of the loss of this spot fluorescence and its treadmilling speed depend on E-cadherin stability in pAJs. This dependence suggests that homeostasis of F-actin in the AJ-BS and AJ-BTs as well as that of cadherin clusters is interconnected, despite the fact that, as indicated above, F-actin in AJ-BT and AJ-BS use different actin polymerization pathways. This complex organization of pAJs may also be relevant to other types of AJs. Indeed, branched actin interconnecting CCCs with actin bundles have been detected in linear AJs (Efimova and Svitkina, 2018), and increased rearward treadmilling induced by the cis E-cadherin mutant was noted in actin bundles connected to E-cadherin–coated substrates (Strale et al., 2015).

Our findings reveal remarkable similarities between AJ-BSs and the dorsal actin bundles linked to FAs. Both types of bundles are enriched with Cnn2 and Cnn3, but not with MyoIIA (Tojkander et al., 2012). They both include relatively stable F-actin, which continuously polymerizes at the junction-attached end of the bundle and then flows rearward. In the case of FAs, the most active polymerization occurs at the interface between the bundle and integrin-associated proteins (Hotulainen and Lappalainen, 2006). The opposite ends of these bundles, distal from junctions, are interconnected with the actomyosin contractile system. Analogous to our observations with pAJs, all proteins within FAs turn over rapidly despite the overall morphological stability of the FA (reviewed in Geiger et al., 2009; Case and Waterman, 2015). Moreover, the adhesive regions of FAs consist of small integrin clusters (reviewed in Changede and Sheetz, 2017), while those in pAJs consist of small cadherin clusters (Indra et al., 2018). Nevertheless, specialized connecting structures analogous to AJ-BTs have not been detected for FAs. It should be noted, however, that F-actin directly associated with FAs is enriched with the same proteins as F-actin in AJ-BTs: vinculin, mena, VASP, and FBLIM1 among them.

Both AJs and FAs function as key force transmission complexes of cells. They sense tensile forces exerted on cells and instruct cell behavior in accordance with these forces (reviewed in Geiger et al., 2009; Huveneers and de Rooij, 2013; Mège and Ishiyama, 2017; Yap et al., 2018). However, our results suggest that AJs sense attached cells using a unique mechanism, which allows them to be rigid enough to transmit force while still being fluid enough to facilitate cell migration. Specifically, the half of a pAJ in one cell senses the dynamics of cadherin and F-actin in the half of the pAJ in the adjacent cell, thus allowing for synchronization of their dynamics. Potentially, this sensing, which we term “dynasensing,” may play a critical role in morphogenesis, for example, by accelerating the movement of particular types of cells within a stable epithelial sheet. The mechanism suggests how mobile cells could instruct stationary cells to form flexible AJs, which, rather than impeding cell motion, could facilitate movement. Such sensing and synchronization of junction dynamics could thus facilitate cell intercalating events during normal morphogenesis or the invasion of malignant cells.

In conclusion, we show that pAJ-linked actin bundles consist of a stable stalk and a dynamic tip. In the tip, short-lived F-actin polymerization-depolymerization cycles are tightly coupled with assembly–disassembly cycles of cadherin clusters. The coupling between E-cadherin and actin turnover allows pAJs to function not only as flexible mechanosensing connectors between the actin cytoskeleton of neighboring cells, but also as sensors and synchronizers of their cytoskeletal dynamics.

STAR[large star]METHODS

LEAD CONTACT AND MATERIALS AVAILABILITY

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Sergey M. Troyanovsky (ude.nretsewhtron@yksvonayort-s). Materials generated in this study are available from the Lead Contact without restrictions.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Cells and transfection

A431 cell line used in this study derived from an epidermal carcinoma. Cells were cultured in DMEM supplemented with 10% Fetal Bovine Serum. Transfection and growth of A431 cells and A431-EcKO cells were done as described in Hong et al. (2011) and Indra et al. (2018). After transfection and antibiotic selection, the cells were sorted for moderate transgene expression by FACS. Importantly, A431-EcKO version of A431 cells were used for expression of the recombinant E-cadherin or its mutants. The expression of the endogenous E-cadherin in these cells was knocked out by CRISPR-Cas9 approach (Indra et al., 2018). To knockout Cfl1 and Aip1 (the name of corresponding gene is WDR1) in A431-EcGFP cells, the Alt-Rtm CRISPR-Cas9 System (IDT, Coralville, IA) was used as it was described previously (Choi et al. (2019). The cells were transfected with an RNA complex consisting of Cfl1-specific or Aip1-specific guide RNAs (5′-tctatgatgcaacctatgag or 5′-ggcgtctccaagatcatcgg designed by software of Broad Institute of Harvard and MIT) and transactivating RNA. The deficient cell clones were then selected through cloning and immunostaining.

METHOD DETAILS

Plasmids

The plasmid encoding E-cadherin and cis-E-cadherin mutant tagged with mGFP and mCH (pRc-EcmGFP and pRcEcmCH) have been reported (Indra et al. (2018). The plasmid containing cis-EcmGFP and P5A-EcmGFP was constructed using pRc-EcmGFP as a template by site directed mutagenesis and verified by sequencing. The plasmid encoding PA-mCH-tagged b-actin was a gift from Dr. Verkhusha (Addgene, plasmid # 31949). pRc-CNN3-mGFP plasmid was generated by using Myc-DDK tagged CNN3 (Origene, RC200488). In brief, full length CNN3 was PCR amplified by flanking unique restriction sites Nhe1 and Not1 at 5′ and 3′ sites respectively and inserted between Nhe1/Not1 sites of pRcCMV-Dn vector.

Immunofluorescence microscopy

For immunofluorescence, cells were fixed with 2% formaldehyde (for 10 min) and permeabilized with 1% Triton X-100 (for 15 min). For Arp2/3 and cortactin staining, the cells were first incubated for 2 min with 1% formaldehyde/0.5%Triton X-100 in cytoskeleton-preserving buffer (80 mM PIPES pH 6.8, 5 mM EGTA, 2 mM MgCl2) described in Heuser and Kirschner (1980) and only then fixed in 2% formaldehyde. In this case the cells were stained without additional permeabilization step. For Cfl staining (Figure 5A), the cells were fixed by iced-cold methanol (5 min) and then acetone (2 min). For confocal microscopy, the cells were cultured 48–64 h on glass-bottom dishes (P35G-1.5; MatTek) and fixed as described above. Immediately before imaging the dishes were filled with 97% glycerol. The images were taken using Nikon Eclipse Ti2-W1-Spinning Disk Perfect Focus microscope equipped with 100x/1.45 NA Plan Apochromat TIRF lens and Hamamatsu Flash 4.0 CMOS Camera. The images were then processed and analyzed using Nikon’s NIS-Element platform version 5.1. SIM images (Fig 2 F) were taken using a Nikon’s Structured Illumination Microscop equipped with Nikon’s Plan Apochromat TIRF 100X oil, NA 1.45 objective lens and Hamamatsu ORCA-Flash4.0 digital CMOS camera. To reconstruct the subresolution structures, the images were processed using NIS-Element v5.0 (Nikon).

The following antibodies were used: mouse antibodies against E-cadherin (clone HECD1 - Zymed Laboratories, South San Francisco, CA), Cfl1 (clone E-8, Santa Cruz Inc., Dallas, TX), VASP (BD Biosciences, San Jose, CA), vinculin and cortactin (clone hVIN-1 and 4F11, Sigma-Aldrich Co., Saint Louis, MO) and rabbit antibodies against MyoIIA (909801 – Biolegend, San Diego, CA), CNN2 and WDR1 (Aip1) (PA5–61878 and PA5–84836 Invitrogen, Carlsbad, CA), CNN3 (ab204365, Abcam, Cambridge MA), mena and FBLIM1 (NBP1–67914, and NBP2–57310 both from Novus Biologicals, Littleton, CO), mCH (5993, BioVision, Mountain View, CA). Three different rabbit antibodies against Arp2/3 complex were used: against ARPC2 and p16 (ab133315 and ab51243, Abcam) and against ARPC2 (07-227, Millipore, Temecula, CA). Specificity of all the antibodies, except anti-mCH and anti-Arp2/3 complex, was tested by a combination of western blotting and specific CRISPR/Cas9 knockout. All three anti-Arp2/3 antibodies yielded the same staining pattern. Secondary antibodies were produced in Donkey (Jackson Immunoresearch Laboratories, West Grove, PA). Alexa Fluor 555-conjugated phalloidin was purchased from Invitrogen.

Live-cell imaging

Cells were imaged (in L-15 media with 10% FBS) by Eclipse Ti microscope (Nikon, Melville, NY) at 37°C controlled with Nikon’s NIS-Elements software. The microscope was equipped with an incubator chamber, a Prime 95B sCMOS camera (Photometrics), halogen and LED light sources and a Plan Apo TIRF DICN2 100x/1.49 lens. The 1×1 binning mode was used in most imaging experiments. At this microscope setting the pixel size was 110 nm. Time-lapse images were taken in both FITC and mCherry filter sets using halogen light that minimize phototoxicity and photobleaching essentially as described earlier (Chen et al., 2015). The mCHPA-actin located at the point of interest was activated by a 50-ms-long exposure to the 405-nm wavelength laser focused to the diffraction-limited spot (~500 nm in diameter). The green and red images were then taken at the indicated intervals (typically 10 s). For FRAP assay a circular ROI with ~500 nm diameter was bleached to approximately 50% of its initial intensity using 40% 405nm laser power.

QUANTIFICATION AND STATISTICAL ANALYSIS

Image quantifications

All images were saved as nd2 files and processed using NIS-Element v5.0 (Nikon) or ImageJ softwares (National Institutes of Health). Line-scan analysis (Figures 2E and and6E)6E) was done using NIS-Element v5.0. Time-lapse images were processed using limited background reduction and denoising functions of NIS-Element. In FRAP and FLIP experiments, the fluorescence intensity of each region was first adjusted by subtracting the background intensity and then normalized to the intensity at the initial time point using NIS Element 5.0. Both red and green channels were treated equally. A single square-size “ROI” was created covering the point of fluorescence bleaching or conversion using ImageJ. The recovery or reduction of fluorescence of the selected ROI was recorded for the entire time lapse images using ImageJ. All graphs, chards and error bars were plotted using GraphPad Prism 8. Fluorescence recovery curves of FRAP analysis (Figures 4A, ,4E,4E, ,5D,5D, and S2) were fitted using Michaelis-Menten equation (Y = Vmax*X/(Km+X)) and Fluorescence loss in photo-activation curves of FLIP analysis (Figures 3D, ,4B,4B, ,4F,4F, and and5E)5E) were fitted using one phase exponential decay (Y = (Y0-NS)*exp(−K*X) + NS). All data were represented as the mean ± standard error of the mean (SEM). All n-values per group are reported in the figure legends. Statistical significance was analyzed using the Student’s two-tailed t test for two groups and ANOVA analysis for more groups. A p value that was less than 0.05 was considered statistically significant.

DATA AND CODE AVAILABILITY

This study did not generate/analyze datasets of codes.

KEY RESOURCES TABLE

REAGENT or RESOURCESOURCEIDENTIFIER
Antibodies
E-cadherin (mouse, monoclonal, clone HECD11)_MilliporeCat# 205601; RRID:AB_211510
Cfl1 (mouse, monoclonal, clone E-8)Santa Cruz BiotechnologyCat# sc-376476, RRID: AB_11150468
VASP (mouse monoclonal, clone 43)BD BiosciencesCat# 610447, RRID:AB_397821
Vinculin (mouse, clone hVIN-1)Sigma-AldrichCat# V9131, RRID:AB_477629
Cortactin (mouse monoclonal, clone 4F11)MilliporeCat# 05–180, RRID:AB_309647
MyoIIA (rabbit polyclonal)BioLegendCat# 909801, RRID:AB_2565100
Calponin-2 (rabbit polyclonal)Thermo Fisher ScientificCat# PA5–61878, RRID:AB_2639249
WDR1 (Aip1) (rabbit polyclonal)Thermo Fisher ScientificCat# PA5–84836, RRID:AB_2791985
Calponin-3 (rabbit polyclonal)AbcamCat# ab204365
Mena (rabbit polyclonal)NovusCat# NBP1–87914, RRID:AB_11017241
Migfilin (FBLIM1) (rabbit polyclonal)novusNBP2–57310
mCherry (rabbit polyclonal)BioVisionCat# 5993–100, RRID:AB_1975001
ARPC2 (rabbit monoclonal)AbcamCat# ab133315
P16 ARC (rabbit monoclonal)AbcamCat# ab51243, RRID:AB_2059963
ARPC2 (rabbit polyclonal)MilliporeCat# 07–227, RRID:AB_11212539
Alexa Fluor 488-Donkey anti-Mouse IgGJackson ImmunoResearch LabsCat# 715-545-150, RRID:AB_2340846
Cy3-Donkey anti-Mouse IgGJackson ImmunoResearch LabsCat# 715-165-150, RRID:AB_2340813
Alexa Fluor 488 Donkey anti-Rabbit IgGJackson ImmunoResearch LabsCat# 711-545-152, RRID:AB_2313584
Cy3-Donkey anti-Rabbit IgGJackson ImmunoResearch LabsLabs Cat# 711-165-152, RRID:AB_2307443
Chemicals, Peptides, and Recombinant Proteins
Phalloidin conjugated with Alexa 555Thermo Fisher ScientificA34055
Critical Commercial Assays
CRISPR-Cas9Alt-Rtm CRISPR-Cas9IDT
Experimental Models: Cell Lines
A431German Cancer Research Center (Heidelberg, FRG)N/A
A431-EcKOIndra et al., 2018N/A
A431-Cnn3-DnThis paperN/A
A431-EcGFPIndra et al., 2018N/A
A431-EcmCHIndra et al., 2018N/A
A431-cis-EcGFPThis paperN/A
A431-P5-AEcGFPThis paperN/A
A431-EcGFP-Cfl1-KOThis paperN/A
A431-EcGFP-Aip1-KOThis paperN/A
Oligonucleotides
transactivating RNA:IDTCat #
Aip1-specific guide RNAs: 5′-ggcgtctccaagatcatcggIDTN/A
Cfl1-specific guide RNAs: 5′-tctatgatgcaacctatgagIDTN/A
Recombinant DNA
PA-mCHSubach et al., 2009Addgene plasmid #31949
pCMVCNN3-MycOriGeneRC200488
pRc-EcmGFPIndra et al., 2018N/A
pRcEcmCHIndra et al., 2018N/A
pRc-CNN3-mGFPThis paperN/A
pRc-P5A-EcmGFPThis paperN/A
pRc-cis-EcmGFPThis paperN/A
Software and Algorithms
NIS-Element 5.0NikonNikon
Prism 8GraphPadGraphPad
FijiIamgeJhttps://imagej.nih.gov/ij/
GPP sgRNA DesignerBROAD Institute GPP Web Portalhttps://portals.broadinstitute.org/gpp/public/analysis-tools/sgrna-design

Highlights

  • Actin bundles connected to adherens junctions consist of a bundle stalk and a tip

  • F-actin in the stalk and tip exhibit different organization and dynamics

  • F-actin and cadherin turnover rates are interdependent

  • Coupling of cadherin and actin dynamics coordinates cells through adherens junctions

Supplementary Material

ACKNOWLEDGMENTS

Sequencing, flow cytometry, and confocal microscopy were performed at the Northwestern University Genetic, Flow Cytometry, and Advanced Microscopy Centers. The work was funded by the National Institutes of Health (grants AR044016 and AR070166 to S.M.T. and GM118584 to L.S.) and the National Science Foundation (grant MCB-1914542 to B.H.).

Footnotes

SUPPLEMENTAL INFORMATION

Supplemental Information can be found online at https://doi.org/10.1016/j.celrep.2020.01.106.

DECLARATION OF INTERESTS

The authors declare no competing interests.

REFERENCES

  • Abe H, Nagaoka R, and Obinata T (1993). Cytoplasmic localization and nuclear transport of cofilin in cultured myotubes. Exp. Cell Res 206, 1–10. [Abstract] [Google Scholar]
  • Bravo-Cordero JJ, Magalhaes MA, Eddy RJ, Hodgson L, and Condeelis J (2013). Functions of cofilin in cell locomotion and invasion. Nat. Rev. Mol. Cell Biol 14, 405–415. [Europe PMC free article] [Abstract] [Google Scholar]
  • Brieher WM, and Yap AS (2013). Cadherin junctions and their cytoskeleton(s). Curr. Opin. Cell Biol 25, 39–46. [Abstract] [Google Scholar]
  • Buckley CD, Tan J, Anderson KL, Hanein D, Volkmann N, Weis WI, Nelson WJ, and Dunn AR (2014). Cell adhesion. The minimal cadherin-catenin complex binds to actin filaments under force. Science 346, 1254211. [Europe PMC free article] [Abstract] [Google Scholar]
  • Case LB, and Waterman CM (2015). Integration of actin dynamics and cell adhesion by a three-dimensional, mechanosensitive molecular clutch. Nat. Cell Biol 17, 955–963. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cavey M, Rauzi M, Lenne PF, and Lecuit T (2008). A two-tiered mechanism for stabilization and immobilization of E-cadherin. Nature 453, 751–756. [Abstract] [Google Scholar]
  • Chan C, Beltzner CC, and Pollard TD (2009). Cofilin dissociates Arp2/3 complex and branches from actin filaments. Curr. Biol 19, 537–545. [Europe PMC free article] [Abstract] [Google Scholar]
  • Changede R, and Sheetz M (2017). Integrin and cadherin clusters: A robust way to organize adhesions for cell mechanics. BioEssays 39, 1–12. [Abstract] [Google Scholar]
  • Chen CS, Hong S, Indra I, Sergeeva AP, Troyanovsky RB, Shapiro L, Honig B, and Troyanovsky SM (2015). α-Catenin-mediated cadherin clustering couples cadherin and actin dynamics. J. Cell Biol 210, 647–661. [Europe PMC free article] [Abstract] [Google Scholar]
  • Choi W, Acharya BR, Peyret G, Fardin MA, Mège RM, Ladoux B, Yap AS, Fanning AS, 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. [Europe PMC free article] [Abstract] [Google Scholar]
  • Choi J, Troyanovsky RB, Indra I, Mitchell BJ, and Troyanovsky SM (2019). Scribble, Erbin, and Lano redundantly regulate epithelial polarity and apical adhesion complex. J. Cell Biol 218, 2277–2293. [Europe PMC free article] [Abstract] [Google Scholar]
  • Chu D, Pan H, Wan P, Wu J, Luo J, Zhu H, and Chen J (2012). AIP1 acts with cofilin to control actin dynamics during epithelial morphogenesis. Development 139, 3561–3571. [Abstract] [Google Scholar]
  • Ciuba K, Hawkes W, Tojkander S, Kogan K, Engel U, Iskratsch T, and Lappalainen P (2018). Calponin-3 is critical for coordinated contractility of actin stress fibers. Sci. Rep 8, 17670. [Europe PMC free article] [Abstract] [Google Scholar]
  • de Beco S, Perney JB, Coscoy S, and Amblard F (2015). Mechanosensitive adaptation of E-cadherin turnover across adherens junctions. PLoS ONE 10, e0128281. [Europe PMC free article] [Abstract] [Google Scholar]
  • Efimova N, and Svitkina TM (2018). Branched actin networks push against each other at adherens junctions to maintain cell-cell adhesion. J. Cell Biol 217, 1827–1845. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fritzsche M, Lewalle A, Duke T, Kruse K, and Charras G (2013). Analysis of turnover dynamics of the submembranous actin cortex. Mol. Biol. Cell 24, 757–767. [Europe PMC free article] [Abstract] [Google Scholar]
  • Geiger B, Spatz JP, and Bershadsky AD (2009). Environmental sensing through focal adhesions. Nat. Rev. Mol. Cell Biol 10, 21–33. [Abstract] [Google Scholar]
  • Guo Z, Neilson LJ, Zhong H, Murray PS, Zanivan S, and Zaidel-Bar R (2014). E-cadherin interactome complexity and robustness resolved by quantitative proteomics. Sci. Signal 7, rs7. [Europe PMC free article] [Abstract] [Google Scholar]
  • Han SP, Gambin Y, Gomez GA, Verma S, Giles N, Michael M, Wu SK, Guo Z, Johnston W, Sierecki E, et al. (2014). Cortactin scaffolds Arp2/3 and WAVE2 at the epithelial zonula adherens. J. Biol. Chem 289, 7764–7775. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hansen SD, Kwiatkowski AV, Ouyang CY, Liu H, Pokutta S, Watkins SC, Volkmann N, Hanein D, Weis WI, Mullins RD, and Nelson WJ (2013). αE-catenin actin-binding domain alters actin filament conformation and regulates binding of nucleation and disassembly factors. Mol. Biol. Cell 24, 3710–3720. [Europe PMC free article] [Abstract] [Google Scholar]
  • Harris TJ, and Tepass U (2010). Adherens junctions: From molecules to morphogenesis. Nat. Rev. Mol. Cell Biol 11, 502–514. [Abstract] [Google Scholar]
  • Harrison OJ, Jin X, Hong S, Bahna F, Ahlsen G, Brasch J, Wu Y, Vendome J, Felsovalyi K, Hampton CM, et al. (2011). The extracellular architecture of adherens junctions revealed by crystal structures of type I cadherins. Structure 19, 244–256. [Europe PMC free article] [Abstract] [Google Scholar]
  • Heuser JE, and Kirschner MW (1980). Filament organization revealed in platinum replicas of freeze-dried cytoskeletons. J. Cell Biol 86, 212–234. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hong S, Troyanovsky RB, and Troyanovsky SM (2011). Cadherin exits the junction by switching its adhesive bond. J. Cell Biol 192, 1073–1083. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hotulainen P, and Lappalainen P (2006). Stress fibers are generated by two distinct actin assembly mechanisms in motile cells. J. Cell Biol 173, 383–394. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hotulainen P, Paunola E, Vartiainen MK, and Lappalainen P (2005). Actin-depolymerizing factor and cofilin-1 play overlapping roles in promoting rapid F-actin depolymerization in mammalian nonmuscle cells. Mol. Biol. Cell 16, 649–664. [Europe PMC free article] [Abstract] [Google Scholar]
  • Huveneers S, and de Rooij J (2013). Mechanosensitive systems at the cadherin-F-actin interface. J. Cell Sci 126, 403–413. [Abstract] [Google Scholar]
  • Indra I, Choi J, Chen CS, Troyanovsky RB, Shapiro L, Honig B, and Troyanovsky SM (2018). Spatial and temporal organization of cadherin in punctate adherens junctions. Proc. Natl. Acad. Sci. USA 115, E4406–E4415. [Europe PMC free article] [Abstract] [Google Scholar]
  • Indra I, Hong S, Troyanovsky R, Kormos B, and Troyanovsky S (2013). The adherens junction: A mosaic of cadherin and nectin clusters bundled by actin filaments. J. Invest. Dermatol 133, 2546–2554. [Europe PMC free article] [Abstract] [Google Scholar]
  • Iwasa JH, and Mullins RD (2007). Spatial and temporal relationships between actin-filament nucleation, capping, and disassembly. Curr. Biol 17, 395–406. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kato A, Kurita S, Hayashi A, Kaji N, Ohashi K, and Mizuno K (2008). Critical roles of actin-interacting protein 1 in cytokinesis and chemotactic migration of mammalian cells. Biochem. J 414, 261–270. [Abstract] [Google Scholar]
  • Kovacs EM, Goodwin M, Ali RG, Paterson AD, and Yap AS (2002). Cadherin-directed actin assembly: E-cadherin physically associates with the Arp2/3 complex to direct actin assembly in nascent adhesive contacts. Curr. Biol 12, 379–382. [Abstract] [Google Scholar]
  • Kovacs EM, Verma S, Ali RG, Ratheesh A, Hamilton NA, Akhmanova A, and Yap AS (2011). N-WASP regulates the epithelial junctional actin cytoskeleton through a non-canonical post-nucleation pathway. Nat. Cell Biol 13, 934–943. [Abstract] [Google Scholar]
  • Lai FP, Szczodrak M, Block J, Faix J, Breitsprecher D, Mannherz HG, Stradal TE, Dunn GA, Small JV, and Rottner K (2008). Arp2/3 complex interactions and actin network turnover in lamellipodia. EMBO J. 27, 982–992. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lappalainen P, and Drubin DG (1997). Cofilin promotes rapid actin filament turnover in vivo. Nature 388, 78–82. [Abstract] [Google Scholar]
  • Lee NK, Fok KW, White A, Wilson NH, O’Leary CJ, Cox HL, Michael M, Yap AS, and Cooper HM (2016). Neogenin recruitment of the WAVE regulatory complex maintains adherens junction stability and tension. Nat. Commun 7, 11082. [Europe PMC free article] [Abstract] [Google Scholar]
  • Leerberg JM, Gomez GA, Verma S, Moussa EJ, Wu SK, Priya R, Hoffman BD, Grashoff C, Schwartz MA, and Yap AS (2014). Tension-sensitive actin assembly supports contractility at the epithelial zonula adherens. Curr. Biol 24, 1689–1699. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li Y, Merkel CD, Zeng X, Heier JA, Cantrell PS, Sun M, Stolz DB, Watkins SC, Yates NA, and Kwiatkowski AV (2019). The N-cadherin interactome in primary cardiomyocytes as defined using quantitative proximity proteomics. J. Cell Sci 132, jcs221606. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mège RM, and Ishiyama N (2017). Integration of cadherin adhesion and cytoskeleton at adherens junctions. Cold Spring Harb. Perspect. Biol 9, a028738. [Europe PMC free article] [Abstract] [Google Scholar]
  • Michael M, and Yap AS (2013). The regulation and functional impact of actin assembly at cadherin cell-cell adhesions. Semin. Cell Dev. Biol 24, 298–307. [Abstract] [Google Scholar]
  • Munsie LN, Desmond CR, and Truant R (2012). Cofilin nuclear-cytoplasmic shuttling affects cofilin-actin rod formation during stress. J. Cell Sci 125, 3977–3988. [Abstract] [Google Scholar]
  • Ono S (2018). Functions of actin-interacting protein 1 (AIP1)/WD repeat protein 1 (WDR1) in actin filament dynamics and cytoskeletal regulation. Biochem Biophys Res Commun 506, 315–322. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pappas DJ, and Rimm DL (2006). Direct interaction of the C-terminal domain of alpha-catenin and F-actin is necessary for stabilized cell-cell adhesion. Cell Commun. Adhes 13, 151–170. [Abstract] [Google Scholar]
  • Scott JA, Shewan AM, den Elzen NR, Loureiro JJ, Gertler FB, and Yap AS (2006). Ena/VASP proteins can regulate distinct modes of actin organization at cadherin-adhesive contacts. Mol. Biol. Cell 17, 1085–1095. [Europe PMC free article] [Abstract] [Google Scholar]
  • Strale PO, Duchesne L, Peyret G, Montel L, Nguyen T, Png E, Tampé R, Troyanovsky S, Hénon S, Ladoux B, and Mège RM (2015). The formation of ordered nanoclusters controls cadherin anchoring to actin and cell-cell contact fluidity. J. Cell Biol 210, 333–346. [Europe PMC free article] [Abstract] [Google Scholar]
  • Subach FV, Patterson GH, Manley S, Gillette JM, Lippincott-Schwartz J, and Verkhusha VV (2009). Photoactivatable mCherry for high-resolution two-color fluorescence microscopy. Nat. Methods 6, 153–159. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tang VW, and Brieher WM (2012). α-Actinin-4/FSGS1 is required for Arp2/3-dependent actin assembly at the adherens junction. J. Cell Biol 196, 115–130. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tojkander S, Gateva G, Husain A, Krishnan R, and Lappalainen P (2015). Generation of contractile actomyosin bundles depends on mechanosensitive actin filament assembly and disassembly. eLife 4, e06126. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tojkander S, Gateva G, and Lappalainen P (2012). Actin stress fibers—assembly, dynamics and biological roles. J. Cell Sci 125, 1855–1864. [Abstract] [Google Scholar]
  • Troyanovsky RB, Sokolov EP, and Troyanovsky SM (2006). Endocytosis of cadherin from intracellular junctions is the driving force for cadherin adhesive dimer disassembly. Mol. Biol. Cell 17, 3484–3493. [Europe PMC free article] [Abstract] [Google Scholar]
  • Van Itallie CM, Tietgens AJ, Aponte A, Fredriksson K, Fanning AS, Gucek M, and Anderson JM (2014). Biotin ligase tagging identifies proteins proximal to E-cadherin, including lipoma preferred partner, a regulator of epithelial cell-cell and cell-substrate adhesion. J. Cell Sci 127, 885–895. [Europe PMC free article] [Abstract] [Google Scholar]
  • Vasioukhin V, Bauer C, Yin M, and Fuchs E (2000). Directed actin polymerization is the driving force for epithelial cell-cell adhesion. Cell 100, 209–219. [Abstract] [Google Scholar]
  • Vendome J, Posy S, Jin X, Bahna F, Ahlsen G, Shapiro L, and Honig B (2011). Molecular design principles underlying β-strand swapping in the adhesive dimerization of cadherins. Nat. Struct. Mol. Biol 18, 693–700. [Europe PMC free article] [Abstract] [Google Scholar]
  • Verma S, Shewan AM, Scott JA, Helwani FM, den Elzen NR, Miki H, Takenawa T, and Yap AS (2004). Arp2/3 activity is necessary for efficient formation of E-cadherin adhesive contacts. J. Biol. Chem 279, 34062–34070. [Abstract] [Google Scholar]
  • Vitriol EA, Wise AL, Berginski ME, Bamburg JR, and Zheng JQ (2013). Instantaneous inactivation of cofilin reveals its function of F-actin disassembly in lamellipodia. Mol. Biol. Cell 24, 2238–2247. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang D, Naydenov NG, Feygin A, Baranwal S, Kuemmerle JF, and Ivanov AI (2016). Actin-depolymerizing factor and Cofilin-1 have unique and overlapping functions in regulating intestinal epithelial junctions and mucosal inflammation. Am. J. Pathol 186, 844–858. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wu SK, Gomez GA, Michael M, Verma S, Cox HL, Lefevre JG, Parton RG, Hamilton NA, Neufeld Z, and Yap AS (2014). Cortical F-actin stabilization generates apical-lateral patterns of junctional contractility that integrate cells into epithelia. Nat Cell Biol 16, 167–178. [Abstract] [Google Scholar]
  • Yamada S, Pokutta S, Drees F, Weis WI, and Nelson WJ (2005). Deconstructing the cadherin-catenin-actin complex. Cell 123, 889–901. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yap AS, Duszyc K, and Viasnoff V (2018). Mechanosensing and mechano-transduction at cell-cell junctions. Cold Spring Harb. Perspect. Biol 10, a028761. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yonemura S, Itoh M, Nagafuchi A, and Tsukita S (1995). Cell-to-cell adherens junction formation and actin filament organization: Similarities and differences between non-polarized fibroblasts and polarized epithelial cells. J. Cell Sci 108, 127–142. [Abstract] [Google Scholar]
  • Zhang J, Betson M, Erasmus J, Zeikos K, Bailly M, Cramer LP, and Braga VM (2005). Actin at cell-cell junctions is composed of two dynamic and functional populations. J. Cell Sci 118, 5549–5562. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/76812469
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/76812469

Article citations


Go to all (20) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.

Funding 


Funders who supported this work.

NIAMS NIH HHS (3)

NIGMS NIH HHS (1)

National Institutes of Health

    National Science Foundation