Arp2/3 complex-nucleated branched actin networks provide the key force necessary for endocytosis. The Arp2/3 complex is activated by nucleation-promoting factors including the Schizosaccharomyces pombe Wiskott–Aldrich syndrome protein (Wsp1) and myosin-1 (Myo1). There are >40 known yeast endocytic proteins with distinct spatial and temporal localizations and functions; however, it is still unclear how these proteins work together to drive endocytosis. Here, we used quantitative live-cell imaging to determine the function of the uncharacterized S. pombe protein Bbc1. We discovered that Myo1 interacts with and recruits Bbc1 to sites of endocytosis. Bbc1 competes with the verprolin Vrp1 for localization to patches and association with Myo1, thus releasing Vrp1 and its binding partner Wsp1 from Myo1. Normally Myo1 remains at the base of the endocytic invagination and Vrp1–Wsp1 internalizes with the endocytic vesicle. However, in the absence of Bbc1, a portion of Vrp1–Wsp1 remains with Myo1 at the base of the invagination, and endocytic structures internalize twice as far. We propose that Bbc1 disrupts a transient interaction of Myo1 with Vrp1 and Wsp1 and thereby limits Arp2/3 complex-mediated nucleation of actin branches at the plasma membrane.

This article has an associated First Person interview with the first author of the paper.

Clathrin-mediated endocytosis (CME) is an essential process conserved across eukaryotes, which allows for the internalization of external materials into a cell. This process is highly regulated with >40 proteins known to assemble into patch structures at sites of endocytosis in a consistent spatial and temporal pattern (Goode et al., 2015; Kovar et al., 2011; McMahon and Boucrot, 2011; Weinberg and Drubin, 2012). How these proteins coordinate their activities to ensure reproducible endocytic patch assembly and efficient endocytosis is an important open question in the field. Although many of the endocytic patch proteins are conserved among species, some are specific to animals or yeast. Bbc1 (also known as Mti1) is a yeast-specific protein that localizes to sites of endocytosis and is thought to function in regulating actin assembly (Kaksonen et al., 2005; Mochida et al., 2002; Picco et al., 2018; Rodal et al., 2003; Sun et al., 2006).

To help overcome membrane tension in yeast and animal cells, endocytosis relies upon the assembly of actin into structures called actin patches, which provide the scaffold and force needed for efficient membrane invagination and scission (Aghamohammadzadeh and Ayscough, 2009; Basu et al., 2014; Boulant et al., 2011; Engqvist-Goldstein and Drubin, 2003; Goode et al., 2015; Kaksonen and Roux, 2018; Kaksonen et al., 2006). The branched actin network at endocytic sites (Collins et al., 2011; Rodal et al., 2005; Young et al., 2004) is assembled by the Arp2/3 complex upon its activation by nucleation-promoting factors (NPFs) (Higgs and Pollard, 2001). Multiple NPFs are present at endocytic sites in fission yeast Schizosaccharomyces pombe (Basu and Chang, 2011; Carnahan and Gould, 2003; Kovar et al., 2011; Sirotkin et al., 2005, 2010) including the Wiskott–Aldrich Syndrome protein (WASp) homolog Wsp1, myosin-1 (Myo1), the EPS15 homolog Pan1, and the SPIN90 homolog Dip1. Homologs of these proteins are also present in budding yeast Saccharomyces cerevisiae, including the WASp Las17 and the myosin-1 proteins Myo3 and Myo5 (Myo3/5) (Boettner et al., 2012; Goode et al., 2015; Weinberg and Drubin, 2012). NPFs share the ability to stimulate the Arp2/3 complex but differ in the strength and mechanism of Arp2/3 complex activation and have additional distinct functions (Galletta et al., 2008; Sirotkin et al., 2005; Sun et al., 2006; Wagner et al., 2013). Specifically, in both budding and fission yeast, WASp is the strongest Arp2/3 complex activator and is considered the primary NPF driving actin assembly for membrane deformation, while myosin-1 is a weak activator that is thought to contribute force via its actin-dependent motor activity (Basu et al., 2014; Berro et al., 2010; Sirotkin et al., 2005; Sun et al., 2006). These diverse NPFs collaborate during endocytosis. In budding yeast, Las17 and Myo3/5 form a WASp–myosin-1 module (Kaksonen et al., 2005; Weinberg and Drubin, 2012) that also includes the F-BAR protein Bzz1 and the WASp-interacting protein (WIP) homolog verprolin (Vrp1), both of which regulate NPF activity in budding and fission yeast (Arasada and Pollard, 2011; Sirotkin et al., 2005; Soulard et al., 2002; Sun et al., 2006). A similar collaboration exists in fission yeast where Myo1 and Wsp1 interact directly (Carnahan and Gould, 2003) or indirectly via Vrp1, which is recruited to patches by Wsp1 but can also bind Myo1 and stimulate its NPF activity (Sirotkin et al., 2005).

The localization and activity of NPFs are regulated to ensure proper actin polymerization. One proposed regulatory component of the budding yeast WASp–myosin-1 module of endocytosis is Bbc1 (Kaksonen et al., 2005; Weinberg and Drubin, 2012). Bbc1 was originally identified in S. cerevisiae as a binding partner of the myosin-1 Myo5 (Mochida et al., 2002) and was shown to mildly inhibit Myo5 NPF activity in vitro (Sun et al., 2006). However, Bbc1 also interacts with Las17 (Tong et al., 2002). Consistent with this interaction, Bbc1, along with Myo5, is recruited to Las17-coated beads but not to Myo5 in comet tails in in vitro reconstitution assays (Michelot et al., 2010). Moreover, a pioneering study by Rodal et al. (2003) demonstrated that Bbc1, in cooperation with adaptor protein Sla1, inhibits Las17 NPF activity in vitro. This work led to a widely accepted model for Bbc1 and Sla1 as dual Las17 inhibitors (Weinberg and Drubin, 2012). Supporting an inhibitory function of Bbc1, bbc1Δ sla1Δ and, to a lesser extent, bbc1Δ cells demonstrate enhanced actin patch assembly (Kaksonen et al., 2005; Picco et al., 2018). The bbc1Δ cells are also characterized by deeper internalizations of endocytic structures (Kaksonen et al., 2005; Picco et al., 2018). However, this depends on the NPF activity of Myo5 but not Las17 (Sun et al., 2006), consistent with identification of Bbc1 as a Myo5 ligand. The behavior and precise localization of Bbc1 at endocytic sites further suggest that Bbc1 interaction with Myo5 may be important for additional, yet to be discovered, functions of Bbc1 in cells. Specifically, S. cerevisiae Bbc1 is recruited to actin patches with timing similar to Myo3/5 (Kaksonen et al., 2005) and, while immuno-electron microscopy (EM) showed that Bbc1 was localized near both Las17 and Myo3/5 in vivo (Idrissi et al., 2008), a recent super-resolution microscopy study placed Bbc1 closer to Myo3/5 than Las17 (Mund et al., 2018). However, the significance of the Bbc1 interaction with myosin-1, aside from the ability of S. cerevisiae Bbc1 to mildly inhibit Myo5 NPF activity in vitro (Sun et al., 2006), remains largely unknown.

Interestingly, the localization pattern of key NPFs differs in budding and fission yeasts. In budding yeast, Myo3/5 and Las17 remain at the base of the invagination and do not internalize with the endocytic vesicle (Kaksonen et al., 2005), except for a small portion detected by immuno-EM (Idrissi et al., 2008) or unless Las17 is overexpressed (Galletta et al., 2008). In contrast, S. pombe Myo1 and Wsp1 show distinct localization and behavior. Myo1 does not internalize with the actin patch and remains at the base of the endocytic invagination, while Wsp1 internalizes with the patch and separates from Myo1 (Sirotkin et al., 2005). This spatial separation of Myo1 and Wsp1 in S. pombe makes it possible to determine whether regulators of actin patch assembly function via association with Myo1 or Wsp1 by following protein dynamics in live cells.

We detected a protein homologous to budding yeast Bbc1/Mti1 in pulldowns of the contractile ring (CR) proteins Cyk3 (Roberts-Galbraith et al., 2010) and Fic1, piquing our interest in this thus far uncharacterized S. pombe protein. Here, we take advantage of distinct behaviors of Myo1 and Wsp1 in S. pombe to reveal a novel mechanism for how Bbc1 regulates actin assembly based on a competition between Bbc1 and Vrp1–Wsp1 for the Myo1 tail.

Bbc1 localizes to the cell division site but not the contractile ring

The protein SPAC23A1.17/Mti1/Bbc1 was identified by mass spectrometry (MS) in affinity purifications of the CR proteins Cyk3 (Roberts-Galbraith et al., 2010) and Fic1 (Table S1), prompting us to investigate localization of this uncharacterized S. pombe protein in dividing cells (Fig. 1). Bbc1–mNeonGreen (mNG) was absent from the CR labeled with mCherry–Cdc15 but was present at the division site in two types of structures: first, in dynamic cortical puncta and, upon completion of CR constriction, also in stable puncta (Fig. 1A–D). Imaging cells expressing Bbc1–mNG, the spindle pole body (SPB) marker Sid4–mNG and the CR marker mCherry–Cdc15 revealed that Bbc1–mNG first appeared in dynamic puncta at the division site 20.9±2.7 min (mean±s.d.) after the separation of SPBs (Fig. 1C). This timing is similar to the time of appearance of endocytic actin patches at the division site (Wu et al., 2006). Imaging of cells expressing Bbc1–mGFP and endocytic actin patch marker Fim1–mCherry confirmed that the dynamic cortical puncta of Bbc1 at the division site are indeed actin patches (Fig. 1E) and stable puncta are distinct from actin patches (Fig. 1F). Bbc1 in stable puncta at new cell tips colocalized with Cdc15 and Cyk3 (Fig. 1G,H), thus explaining isolation of Bbc1 with these proteins in affinity purifications.

Fig. 1.

Bbc1 localizes to the cell division site but not the CR. (A) Live-cell images of mCherry (mCh)–Cdc15 (magenta), Bbc1–mNeonGreen (mNG) and Sid4–mNG (green) at different cell cycle stages. Arrows indicate Bbc1 cortical puncta. BF, bright-field images. Scale bar: 5 µm. (B) Images at 4-min intervals of cells expressing mCherry–Cdc15 (magenta), Bbc1–mNG and Sid4–-mNG (green). Scale bar: 5 µm. (C) Timeline showing detection of Bbc1 at the division site. Time 0 was defined as the time of SPB separation. Error bars represent s.e.m.; n=23 cells. (D) End-on images at 4-min intervals of cells expressing the CR marker Rlc1–mCherry (magenta) and Bbc1–mNG (green). Scale bars: 2 µm. (E,F) Images at 2 seconds per frame (SPF) of Bbc1–mGFP (green) and Fim1–mCherry (magenta) at the division site (boxed in teal) at the (E) beginning and (F) end of CR constriction. Arrowheads mark dynamic patches and arrows mark stable puncta. Scale bars, 2 µm. (G,H) Colocalization of Bbc1, Cdc15, and Cyk3 in stable puncta. (G) Time series of images of Bbc1–mGFP and mCherry–Cdc15 in a stable punctum. (H) Bbc1–mGFP (mG) and Cyk3–mCherry in a stable punctum. Scale bars: whole cells, 2 µm; montages and punctum, 1 µm.

Fig. 1.

Bbc1 localizes to the cell division site but not the CR. (A) Live-cell images of mCherry (mCh)–Cdc15 (magenta), Bbc1–mNeonGreen (mNG) and Sid4–mNG (green) at different cell cycle stages. Arrows indicate Bbc1 cortical puncta. BF, bright-field images. Scale bar: 5 µm. (B) Images at 4-min intervals of cells expressing mCherry–Cdc15 (magenta), Bbc1–mNG and Sid4–-mNG (green). Scale bar: 5 µm. (C) Timeline showing detection of Bbc1 at the division site. Time 0 was defined as the time of SPB separation. Error bars represent s.e.m.; n=23 cells. (D) End-on images at 4-min intervals of cells expressing the CR marker Rlc1–mCherry (magenta) and Bbc1–mNG (green). Scale bars: 2 µm. (E,F) Images at 2 seconds per frame (SPF) of Bbc1–mGFP (green) and Fim1–mCherry (magenta) at the division site (boxed in teal) at the (E) beginning and (F) end of CR constriction. Arrowheads mark dynamic patches and arrows mark stable puncta. Scale bars, 2 µm. (G,H) Colocalization of Bbc1, Cdc15, and Cyk3 in stable puncta. (G) Time series of images of Bbc1–mGFP and mCherry–Cdc15 in a stable punctum. (H) Bbc1–mGFP (mG) and Cyk3–mCherry in a stable punctum. Scale bars: whole cells, 2 µm; montages and punctum, 1 µm.

To gain insight into the physiological role of Bbc1, we deleted bbc1+ and examined genetic interactions of bbc1Δ and sensitivity of bbc1Δ cells to a variety of stresses. The bbc1Δ cells were sensitive to benomyl and latrunculin A (Fig. S1A) and showed negative interactions with fim1Δ, myo1Δ, zds1Δ and mid1Δ (Fig. S1B), implicating Bbc1 in endocytosis, cell wall integrity and cytokinesis (Chang et al., 1996; Sirotkin et al., 2005; Skau and Kovar, 2010; Sohrmann et al., 1996; Yakura et al., 2006). However, bbc1Δ cells were morphologically normal and had no apparent cytokinesis defects, and Bbc1-overexpressing cells had a normal septation index (Fig. S1C). Given that S. pombe Bbc1 localizes to actin patches and co-purifies with actin patch components Myo1, Pan1 and Cdc15 in affinity purifications (Table S1), we then investigated the role of S. pombe Bbc1 in endocytic actin patches.

Bbc1 localizes to the base of the endocytic invagination in a Myo1-dependent manner

We confirmed, by colocalization analysis with actin patch components Fim1 and Pan1, that in interphase cells, as during cell division, Bbc1 localizes to both actin patches and actin-free puncta (Fig. 2A,B; Fig. S2A). During interphase, Bbc1 colocalizes with Cdc15 in actin patches and also in a subpopulation of stable puncta (Fig. 2C; Fig. S2B,C). During cell division, Cdc15 localizes to the CR and is absent from actin patches (Arasada and Pollard, 2011; Carnahan and Gould, 2003; Wu et al., 2006).

Fig. 2.

Bbc1 localizes to the base of the endocytic invagination in a Myo1-dependent manner. (A–C) Time-lapse images of (A) Bbc1–mGFP (green) and Fim1–mCherry (magenta), (B) Bbc1–mNG (green) and Pan1–mCherry (magenta), and (C) Bbc1–mNG (green) and Cdc15–mCherry (magenta) at 2-s (A,B) or 1-s (C) intervals in boxed regions of single live interphase cells (left panels). SPF, seconds per frame. In A, arrowheads indicate an actin patch in a region boxed in teal and arrows indicate a stable punctum in a region boxed in yellow. In B and C, arrows mark protein appearances in endocytic actin patches. Scale bars: cell panel, 2 µm, montages, 1 µm (A); cell panel, 5 µm; montages, 2 µm (B); cell panel and montages, 5 µm (C). (D) Average time courses of the number of molecules of mGFP-tagged Bbc1, Cdc15, Wsp1 and Myo1, Fim1-mCherry fluorescence intensity, and distance traveled by each marker in endocytic actin patches. Time courses are aligned to the start of Fim1–mCherry patch movement (time zero). n=6–9 patches per strain. (E) Maximum intensity projections of time series of images acquired at 1-s interval for 60 s in a single confocal section through the middle of the WT, wsp1Δ, and myo1Δ cells expressing Bbc1–mGFP (left) or Cdc15–mGFP (right). Scale bar: 2 μm. Yellow arrowheads indicate stable puncta. (F) Average time courses of intensities and bar graphs of peak intensities of Bbc1–mGFP and Cdc15–mGPF in endocytic patches in WT (blue) and wsp1Δ (red) cells. Time courses are aligned to peak patch intensity (t=0). P-values represent statistical significance as determined by t-test. N indicates number of patches analyzed. Error bars represent s.d.

Fig. 2.

Bbc1 localizes to the base of the endocytic invagination in a Myo1-dependent manner. (A–C) Time-lapse images of (A) Bbc1–mGFP (green) and Fim1–mCherry (magenta), (B) Bbc1–mNG (green) and Pan1–mCherry (magenta), and (C) Bbc1–mNG (green) and Cdc15–mCherry (magenta) at 2-s (A,B) or 1-s (C) intervals in boxed regions of single live interphase cells (left panels). SPF, seconds per frame. In A, arrowheads indicate an actin patch in a region boxed in teal and arrows indicate a stable punctum in a region boxed in yellow. In B and C, arrows mark protein appearances in endocytic actin patches. Scale bars: cell panel, 2 µm, montages, 1 µm (A); cell panel, 5 µm; montages, 2 µm (B); cell panel and montages, 5 µm (C). (D) Average time courses of the number of molecules of mGFP-tagged Bbc1, Cdc15, Wsp1 and Myo1, Fim1-mCherry fluorescence intensity, and distance traveled by each marker in endocytic actin patches. Time courses are aligned to the start of Fim1–mCherry patch movement (time zero). n=6–9 patches per strain. (E) Maximum intensity projections of time series of images acquired at 1-s interval for 60 s in a single confocal section through the middle of the WT, wsp1Δ, and myo1Δ cells expressing Bbc1–mGFP (left) or Cdc15–mGFP (right). Scale bar: 2 μm. Yellow arrowheads indicate stable puncta. (F) Average time courses of intensities and bar graphs of peak intensities of Bbc1–mGFP and Cdc15–mGPF in endocytic patches in WT (blue) and wsp1Δ (red) cells. Time courses are aligned to peak patch intensity (t=0). P-values represent statistical significance as determined by t-test. N indicates number of patches analyzed. Error bars represent s.d.

Quantitative imaging of strains combining Fim1–mCherry with mGFP-tagged Bbc1 or other patch proteins provided detailed accounting of Bbc1 dynamics in patches relative to Myo1, Wsp1 and Cdc15, patch markers with known dynamics (Arasada and Pollard, 2011; Sirotkin et al., 2010). We determined the time courses of the numbers of molecules of these proteins in actin patches (Fig. 2D; Fig. S2D–H) based on previously measured numbers of actin patch proteins (Sirotkin et al., 2010). Time courses of the numbers of molecules were aligned to the time when patches marked by Fim1–mCherry first moved (time zero), which corresponds to the initiation of actin patch internalization (Berro and Pollard, 2014). S. pombe Bbc1 appeared in actin patches 4–6 s before the start of actin patch internalization, reached peak number of 48 molecules at −2 s, and quickly dissipated 2–6 s after time zero. Cdc15 showed similar behavior, appearing at −4 to −2 s, peaking at 74 molecules at time zero, and disappearing 2–4 s later, comparable to previous measurements (Arasada and Pollard, 2011). Both Myo1 and Wsp1 are present in excess over Bbc1 and Cdc15, reaching peaks of 359 and 155 molecules, respectively, at −2 s. In contrast to Wsp1, which in S. pombe internalizes with the actin patch, neither Bbc1 nor, as previously reported (Arasada and Pollard, 2011), Cdc15 internalized with actin patches. This behavior of Bbc1 and Cdc15 resembles that of Myo1, suggesting Bbc1 and Cdc15 function with Myo1 at the base of the endocytic invagination.

To determine whether Myo1 or Wsp1 play a role in Bbc1 localization in patches, we examined localization of Bbc1–mGFP in myo1Δ and wsp1Δ cells using Cdc15, which is known to depend on Myo1 for localization to actin patches (Arasada and Pollard, 2011), as a control (Fig. 2E,F). We found that both Bbc1–mGFP and Cdc15–mGFP failed to localize to dynamic cortical patches in myo1Δ cells (Fig. 2E) and instead localized exclusively in stable cortical puncta (Fig. S2A,B). In contrast, in the absence of Wsp1, both Bbc1–mGFP and Cdc15–mGFP continued to localize in dynamic patches (Fig. 2E), albeit with slower dynamics (Fig. 2F) typical for endocytic patches in wsp1Δ cells (Basu et al., 2014). Interestingly, while Cdc15 accumulated in patches to the same level in wsp1Δ as in wild-type cells, the peak level of Bbc1 in patches in wsp1Δ cells was increased by ∼25% (Fig. 2F). Thus, Bbc1 is recruited to patches by Myo1 and may compete with other ligands for binding Myo1.

Myo1 recruits Bbc1 through the TH2 and SH3 domains

To determine which part of Myo1 is responsible for recruiting Bbc1 and Cdc15 to patches, we examined the ability of Bbc1 and Cdc15 to localize to patches marked by Fim1–mCherry in strains featuring a series of Myo1 tail truncations using two-color imaging (Fig. 3A; Fig. S3A). We uncovered that the Myo1 TH2 and SH3 domains are both required for Bbc1 patch recruitment (Fig. 3A). In contrast, the Myo1 TH2 and SH3 domains contribute but are not required for Cdc15 recruitment. When the Myo1 TH2 domain, SH3 domain or both were deleted (Fig. S3A), Cdc15 continued to localize to some of the patches, albeit at decreased levels. This is consistent with previous mapping of Myo1–Cdc15 interaction to the TH1 domain in addition to a portion of the TH2 domain (Carnahan and Gould, 2003).

Fig. 3.

The Myo1 SH3 domain is necessary to recruit Bbc1. (A) A schematic of the Myo1 protein domains with relevant binding partners and images at 2 seconds per frame (SPF) from colocalization analysis of Bbc1–mGFP (green) and Fim1–mCherry (magenta) in patches (arrowheads) in WT and the indicated Myo1 tail domain deletion strains. Numbers show the fraction of Fim1 patches that also contained Bbc1. Scale bars: whole cells, 2 μm; montages, 1 μm. (B) Colocalization of Bbc1–mGFP (green) with a Ppc89–mCherry–23A (magenta) aggregate in a single cell (outlined). A line scan through the aggregate confirms colocalization. (C,D) Representative images and line scans from colocalization analysis of depicted (C) Ppc89–mCherry–23A or (D) Ppc89–mCherry–3A protein constructs (top) expressed from plasmids in WT cells or in cells expressing mGFP-tagged Bbc1, Cdc15, Vrp1, Wsp1 and Sla1 from endogenous loci. The graphs show single line scans of mCherry and mGFP intensities along the lines across aggregates indicated by white lines on merged mCherry (magenta) and mGFP (green) images. Numbers indicate percent colocalization; n indicates number of aggregates analyzed for each condition. Scale bars: 1 μm.

Fig. 3.

The Myo1 SH3 domain is necessary to recruit Bbc1. (A) A schematic of the Myo1 protein domains with relevant binding partners and images at 2 seconds per frame (SPF) from colocalization analysis of Bbc1–mGFP (green) and Fim1–mCherry (magenta) in patches (arrowheads) in WT and the indicated Myo1 tail domain deletion strains. Numbers show the fraction of Fim1 patches that also contained Bbc1. Scale bars: whole cells, 2 μm; montages, 1 μm. (B) Colocalization of Bbc1–mGFP (green) with a Ppc89–mCherry–23A (magenta) aggregate in a single cell (outlined). A line scan through the aggregate confirms colocalization. (C,D) Representative images and line scans from colocalization analysis of depicted (C) Ppc89–mCherry–23A or (D) Ppc89–mCherry–3A protein constructs (top) expressed from plasmids in WT cells or in cells expressing mGFP-tagged Bbc1, Cdc15, Vrp1, Wsp1 and Sla1 from endogenous loci. The graphs show single line scans of mCherry and mGFP intensities along the lines across aggregates indicated by white lines on merged mCherry (magenta) and mGFP (green) images. Numbers indicate percent colocalization; n indicates number of aggregates analyzed for each condition. Scale bars: 1 μm.

To determine whether S. pombe Bbc1 can associate with the Myo1 tail, we developed an in vivo binding assay since Bbc1 is a 170 kDa proline-rich protein, which hinders its purification and characterization in vitro. We fused the C-terminal domain of the SPB protein Ppc89 (Rosenberg et al., 2006) to mCherry and to Myo1 tail fragments consisting of the TH2, SH3 and LCA domains (Ppc89–mCherry–23A), the SH3 and LCA domains (Ppc89–mCherry–3A), or the LCA domain alone (Ppc89–mCherry–LCA). These constructs were introduced into cells expressing mGFP-tagged Bbc1, other known Myo1 tail ligands Vrp1, Wsp1 and Cdc15 (Carnahan and Gould, 2003; Sirotkin et al., 2005), or Sla1, which is not known to interact with Myo1. Rather than localizing to SPBs as might have been expected, each construct formed aggregates within the cytoplasm. These were still useful, however, to determine whether mGFP-tagged proteins were recruited by mCherry-marked Myo1 fragments (Fig. 3B). With the exception of Sla1, all examined proteins relocalized to the Ppc89–mCherry–23A puncta (Fig. 3C). However, only Bbc1, Vrp1 and Wsp1 relocalized to the Ppc89–mCherry–3A puncta (Fig. 3D). Neither Bbc1 nor Vrp1 relocalized to the Ppc89–mCherry–LCA aggregates, indicating that the Myo1 SH3 domain alone is sufficient to relocalize both Bbc1 and Vrp1 (Fig. S3B). The binding of Cdc15 to the TH2–SH3–LCA but not SH3–LCA Myo1 construct is consistent with Cdc15 interacting with the TH1 and TH2 domains (Carnahan and Gould, 2003) and suggests that SH3 domain helps recruit Cdc15 to patches via indirect interactions. In contrast, the Myo1 SH3 domain is sufficient for interaction with Bbc1 and together with the TH2 domain is necessary for Bbc1 localization to patches.

Vrp1 competes with Bbc1 for localization to patches and helps retain Myo1 in patches

Since the Myo1 SH3 domain is necessary for recruitment of Bbc1 to patches but also interacts with Vrp1 in vivo (Fig. 3D) and in vitro (Sirotkin et al., 2005), we tested whether Vrp1 and Bbc1 compete for localization to patches in cells. We measured accumulation of Bbc1–mGFP in patches in vrp1Δ cells and Vrp1–mGFP in bbc1Δ cells versus wild-type cells, expecting increased accumulation of Bbc1 or Vrp1 in patches in the absence of its potential competitor. Indeed, while whole-cell intensities were unchanged (Fig. S4A,B), Bbc1 patch accumulation was increased by 40% in the absence of Vrp1 (Fig. 4A), and Vrp1 patch accumulation was increased by 51% in the absence of Bbc1 (Fig. 4B). This competition mechanism may also explain the modest 25% increase in the Bbc1 level in patches observed in wsp1Δ cells (Fig. 2F) since Wsp1 recruits Vrp1 to patches (Sirotkin et al., 2005).

Fig. 4.

Bbc1 and Vrp1 compete for Myo1 and modulate NPF accumulation in patches. (A–D) Average time courses of normalized intensities and bar graphs of average normalized peak intensities of (A) Bbc1–mGFP in patches in WT (blue) and vrp1Δ (red) cells, (B) Vrp1–mGFP in patches in WT (blue) and bbc1Δ (green) cells, (C) mGFP–Myo1 in patches in WT (blue), bbc1Δ (red), vrp1Δ (green), bbc1Δ vrp1Δ (purple) cells, and (D) GFP-tagged Myo1 in patches in WT (blue) and bbc1Δ (red) cells. Time courses are aligned (t=0) and normalized to the WT peak intensity. Myo1 patch dynamics were measured in WT (blue) or bbc1Δ (red) cells expressing Myo1–GFP (Sets 1 and 3), Myo1–GFP and LifeAct–mCherry (Set 2), or mGFP-Myo1 (Set 4) and normalized peak intensities for individual patches from all datasets were combined and averaged. Error bars represent s.d. N indicates number of patches analyzed. P-values represent statistical significance as determined by Student's t-test (A,B,D) or ANOVA with Tukey's post-hoc test (C). (E) A schematic of Bbc1 competition with Vrp1 and Wsp1 for the Myo1 SH3 domain. (F–H) Average peak-aligned time courses of intensities and distances traveled by (F) Vrp1–mGFP, (G) mGFP–Myo1 or (H) mGFP–Wsp1 in endocytic patches in WT (blue) or Bbc1-overexpressing (O.E., red) cells. n=6–8 patches per condition.

Fig. 4.

Bbc1 and Vrp1 compete for Myo1 and modulate NPF accumulation in patches. (A–D) Average time courses of normalized intensities and bar graphs of average normalized peak intensities of (A) Bbc1–mGFP in patches in WT (blue) and vrp1Δ (red) cells, (B) Vrp1–mGFP in patches in WT (blue) and bbc1Δ (green) cells, (C) mGFP–Myo1 in patches in WT (blue), bbc1Δ (red), vrp1Δ (green), bbc1Δ vrp1Δ (purple) cells, and (D) GFP-tagged Myo1 in patches in WT (blue) and bbc1Δ (red) cells. Time courses are aligned (t=0) and normalized to the WT peak intensity. Myo1 patch dynamics were measured in WT (blue) or bbc1Δ (red) cells expressing Myo1–GFP (Sets 1 and 3), Myo1–GFP and LifeAct–mCherry (Set 2), or mGFP-Myo1 (Set 4) and normalized peak intensities for individual patches from all datasets were combined and averaged. Error bars represent s.d. N indicates number of patches analyzed. P-values represent statistical significance as determined by Student's t-test (A,B,D) or ANOVA with Tukey's post-hoc test (C). (E) A schematic of Bbc1 competition with Vrp1 and Wsp1 for the Myo1 SH3 domain. (F–H) Average peak-aligned time courses of intensities and distances traveled by (F) Vrp1–mGFP, (G) mGFP–Myo1 or (H) mGFP–Wsp1 in endocytic patches in WT (blue) or Bbc1-overexpressing (O.E., red) cells. n=6–8 patches per condition.

Next, we examined the effects of competition between Bbc1 and Vrp1 on recruitment of Myo1 to patches by measuring Myo1 levels in patches in the absence of Bbc1, Vrp1 or both proteins (Fig. 4C). Myo1 accumulation in patches was reduced by 20–30% in vrp1Δ and bbc1Δ vrp1Δ cells, suggesting that Vrp1 helps retain Myo1 in patches even though these proteins differ in their patch dynamics (Sirotkin et al., 2005). In contrast, Myo1 accumulation was mildly increased in bbc1Δ cells. This slight increase, 15% on average, was reproducible across multiple experiments (Fig. 4D) and can be explained if Myo1 association with Vrp1 is favored in the absence of Bbc1 (Fig. 4E), thus prolonging Myo1 retention in patches.

Verprolins bind long-tailed myosin-1 and WASp homologs through two distinct domains, a proline-rich region and the C-terminal WASp-binding domain, respectively (Anderson et al., 1998; Carnahan and Gould, 2003; Naqvi et al., 1998; Ramesh et al., 1997; Sirotkin et al., 2005). Therefore, S. pombe Vrp1 can bridge Myo1–Wsp1 association and may help retain both Myo1 and Wsp1 in patches (Fig. 4E). We hypothesized that increased interaction of Bbc1 with Myo1 upon Bbc1 overexpression will disrupt this bridging interaction and decrease accumulation of Vrp1, Myo1 and Wsp1 in patches. Indeed, the levels of Vrp1, Myo1 and Wsp1 were significantly reduced in Bbc1 overexpressing cells (Fig. 4F–H, Fig. S4C,D). Collectively, these data support the competitive binding of Bbc1 and Vrp1 to Myo1 in cells, with Vrp1 promoting and Bbc1 reducing accumulation of Myo1 in patches.

Bbc1 and Sla1 inhibit assembly of NPFs and actin at endocytic sites

Actin patch assembly is enhanced in S. cerevisiae cells lacking Bbc1 (Picco et al., 2018) and to an even greater degree in cells lacking both Bbc1 and the adaptor protein Sla1 (Kaksonen et al., 2005). To test whether inhibition of actin assembly by Sla1 and Bbc1 is conserved in S. pombe, we examined the effects of bbc1Δ alone, sla1Δ alone or a combination of bbc1Δ and sla1Δ on accumulation in patches of F-actin labeled with Fim1–mGFP, Crn1–GFP or LifeAct–mCherry (Huang et al., 2012; Pelham and Chang, 2001; Sirotkin et al., 2010) and NPFs Myo1, Wsp1, Vrp1 and Pan1 (Fig. 5; Fig. S5).

Fig. 5.

Bbc1 and Sla1 together limit accumulation of NPFs and actin in patches. (A–F) Average time courses of normalized intensities and distances traveled, and bar graphs of average normalized peak intensities of (A) Myo1–GFP, (B) Vrp1–mGFP, (C) Pan1–GFP, (D) mGFP–Wsp1, (E) Fim1–mGFP in patches in WT (blue), bbc1Δ (red), sla1Δ (green), and bbc1Δ sla1Δ (purple) cells, and (F) mGFP–Wsp1 in bbc1Δ sla1Δ bzz1Δ (red), bbc1Δ sla1Δ vrp1Δ (green), bbc1Δ sla1Δ (purple), and WT (blue) cells. Dashed lines in A,B,D and F represent the fluorescence intensities and distance traveled by portions of patches exhibiting unusual behavior: (A) internalizing portions of Myo1 patches or (B,D,F) basal portions of splitting Wsp1 and Vrp1 patches. Time courses are aligned (t=0) and normalized to the WT peak intensity. Error bars represent s.d. N indicates number of patches analyzed. P-values represent statistical significance as determined by ANOVA with Tukey's post-hoc test.

Fig. 5.

Bbc1 and Sla1 together limit accumulation of NPFs and actin in patches. (A–F) Average time courses of normalized intensities and distances traveled, and bar graphs of average normalized peak intensities of (A) Myo1–GFP, (B) Vrp1–mGFP, (C) Pan1–GFP, (D) mGFP–Wsp1, (E) Fim1–mGFP in patches in WT (blue), bbc1Δ (red), sla1Δ (green), and bbc1Δ sla1Δ (purple) cells, and (F) mGFP–Wsp1 in bbc1Δ sla1Δ bzz1Δ (red), bbc1Δ sla1Δ vrp1Δ (green), bbc1Δ sla1Δ (purple), and WT (blue) cells. Dashed lines in A,B,D and F represent the fluorescence intensities and distance traveled by portions of patches exhibiting unusual behavior: (A) internalizing portions of Myo1 patches or (B,D,F) basal portions of splitting Wsp1 and Vrp1 patches. Time courses are aligned (t=0) and normalized to the WT peak intensity. Error bars represent s.d. N indicates number of patches analyzed. P-values represent statistical significance as determined by ANOVA with Tukey's post-hoc test.

In agreement with the previous observations (Fig. 4), the loss of Bbc1 alone resulted in a mild increase in the accumulation of Myo1 in patches and a notable increase in accumulation of Vrp1 in patches but had no effect on accumulation of Pan1–GFP in patches (Fig. 5A–C; Fig. S5A,B). In contrast to the increased accumulation of WASp Las17 and actin in patches in S. cerevisiae bbc1Δ cells (Kaksonen et al., 2005; Picco et al., 2018), the peak levels and lifetimes in patches for mGFP–Wsp1 and F-actin markers Fim1–mGFP and Crn1-GFP were unaltered in S. pombe bbc1Δ cells (Fig. 5D,E; Fig. S5C,D). Likewise, even though F-actin levels observed with LifeAct–mCherry were inconsistent between experiments, the mean intensities across multiple experiments indicated no change in actin levels in the absence of Bbc1 (Fig. S5E,F). This observation also suggests that Bbc1 may play a role in suppressing variability of F-actin levels in patches.

Examination of sla1Δ cells revealed only modest increases in accumulation of all markers except Pan1. Compared to wild-type cells, peak patch intensities of Myo1–GFP, Vrp1–mGFP, mGFP–Wsp1, Fim1–mGFP and LifeAct–mCherry in sla1Δ cells increased by 28%, 9%, 21%, 30% and 11%, respectively (Fig. 5A,B,D,E; Fig. S5E), while the peak intensity of Pan1–GFP increased by 125% (Fig. 5C). In addition, Pan1 patch lifetime was significantly increased from 35 s to 64 s. The reasons for these dramatic effects of sla1Δ on Pan1 are not clear but a similar increase of Pan1 patch lifetime in sla1Δ cells has been reported in S. cerevisiae (Kaksonen et al., 2005).

Combining bbc1Δ with sla1Δ resulted in greatly increased accumulation of NPFs and F-actin in patches. Compared to wild-type cells, in sla1Δ bbc1Δ cells, Myo1–GFP, Vrp1–mGFP, Pan1–GFP, mGFP–Wsp1, Fim1–mGFP and LifeAct–mCherry peak patch intensities increased by 91%, 146%, 242%, 51–73%, 57% and 120%, respectively (Fig. 5; Fig. S5E). The same trends were observed with N-terminally tagged Myo1, but to a lesser extent (Fig. S5A). Combining bbc1Δ sla1Δ with vrp1Δ, but not with bzz1Δ, reduced Wsp1 accumulation in patches, indicating that Vrp1 helps retain Wsp1 in patches (Fig. 5F). The synergistic rather than additive effects of sla1Δ and bbc1Δ on assembly of NPFs and actin into patches suggest that S. pombe Sla1 and Bbc1 play redundant roles in constraining actin patch assembly.

Myo1 partially internalizes with actin patches in the absence of Sla1

In addition to increased accumulation of Myo1 in patches, sla1Δ and bbc1Δ sla1Δ cells displayed an unusual behavior of Myo1 at the endocytic sites. In wild-type and bbc1Δ cells, Myo1 always remained at the base of endocytic invaginations and did not internalize with vesicles (Sirotkin et al., 2005) (Fig. 6A). In contrast, in 6% of Myo1–mGFP patches in sla1Δ cells, a small portion of Myo1 moved into the cytoplasm, presumably in association with the tip of the endocytic invagination or endocytic vesicle, while a larger portion of Myo1 remained at the base (Fig. 6A,B). This partial internalization of Myo1, observed as splitting of the Myo1 signal, was significantly more pronounced in bbc1Δ sla1Δ cells where the percentage of Myo1 patches exhibiting this behavior increased from 6% to 64% (Fig. 6A,B). We suspect that the Myo1 splitting events were more evident in bbc1Δ sla1Δ cells because, compared to sla1Δ alone, Myo1 patches were brighter and internalized two times farther (Fig. 6C).

Fig. 6.

Myo1 partially internalizes in the absence of Sla1, and Wsp1 and Vrp1 patches split in the absence of Bbc1. (A) Time lapse montages at 1 second per frame (SPF) showing mGFP–Myo1 patches partially internalizing in sla1Δ and bbc1Δ sla1Δ but not in bbc1Δ and WT cells. Arrowheads mark instances of Myo1 patch partial internalization. Scale bar: 1 μm. (B) Quantification of the percentage of Myo1 patches partially internalizing (orange) and not internalizing (normal, gray). *P<0.05 for the difference from sla1Δ (χ-squared test). (C) Bar graphs of the distance traveled by the internalized portion of mGFP–Myo1 patches in sla1Δ and bbc1Δ sla1Δ cells compared to Sla1-mGFP patches in WT cells. Error bars represent s.d. P-values represent statistical significance as determined by ANOVA with Tukey's post-hoc test. (D–F) Single confocal sections through the middle of the cells and (D′–F′) corresponding time lapse montages at 2 SPF showing (D,D′) localization of Myo1–GFP (green) at the base and the tip of the actin plume visualized with LifeAct–mCherry (magenta) in a bbc1Δ sla1Δ cell, (E,E′) mGFP–Myo1 in actin plumes in a sla1Δ cell, and (F,F′) mGFP–Myo1 in actin plumes in a bbc1Δ sla1Δ cell. Insets in D show examples of Myo1–GFP at the tip and kink in the actin plume (arrowheads). Other arrowheads in D–F indicate Myo1 spots shown on montages in D′–F′. Scale bars: 2 μm (D–F, D′–F′); 1 μm (insets in D). (G–H) Montages of time lapse images at 1 SPF of (G) mGFP–Wsp1 and (H) Vrp1–mGFP patches splitting in bbc1Δ and bbc1Δ sla1Δ cells and not splitting in sla1Δ and WT cells. Scale bar: 1 μm. Arrowheads indicate instances of patch splitting. (I,J) Quantification of the percentage of events when (I) mGPF–Wsp1 or (J) Vrp1–mGFP patches internalized normally (blue), remained immobile (black), or split in two (red) in WT, bbc1Δ, sla1Δ and bbc1Δ sla1Δ cells. *P<0.001 for the difference from bbc1Δ (χ-squared test). (K) Diagram of proposed interactions of the Myo1 SH3 domain. (L,M) Effects of (L) vrp1Δ or bzz1Δ and (M) myo1ΔSH3-LCA (myo1Δ3CA) on the percentage of events when mGPF–Wsp1 patches internalized normally (blue), remained immobile (black) or split in two (red) in bbc1Δ sla1Δ cells. *P<0.001 for the difference from bbc1Δ sla1Δ (χ-squared test). N indicates number of patches analyzed.

Fig. 6.

Myo1 partially internalizes in the absence of Sla1, and Wsp1 and Vrp1 patches split in the absence of Bbc1. (A) Time lapse montages at 1 second per frame (SPF) showing mGFP–Myo1 patches partially internalizing in sla1Δ and bbc1Δ sla1Δ but not in bbc1Δ and WT cells. Arrowheads mark instances of Myo1 patch partial internalization. Scale bar: 1 μm. (B) Quantification of the percentage of Myo1 patches partially internalizing (orange) and not internalizing (normal, gray). *P<0.05 for the difference from sla1Δ (χ-squared test). (C) Bar graphs of the distance traveled by the internalized portion of mGFP–Myo1 patches in sla1Δ and bbc1Δ sla1Δ cells compared to Sla1-mGFP patches in WT cells. Error bars represent s.d. P-values represent statistical significance as determined by ANOVA with Tukey's post-hoc test. (D–F) Single confocal sections through the middle of the cells and (D′–F′) corresponding time lapse montages at 2 SPF showing (D,D′) localization of Myo1–GFP (green) at the base and the tip of the actin plume visualized with LifeAct–mCherry (magenta) in a bbc1Δ sla1Δ cell, (E,E′) mGFP–Myo1 in actin plumes in a sla1Δ cell, and (F,F′) mGFP–Myo1 in actin plumes in a bbc1Δ sla1Δ cell. Insets in D show examples of Myo1–GFP at the tip and kink in the actin plume (arrowheads). Other arrowheads in D–F indicate Myo1 spots shown on montages in D′–F′. Scale bars: 2 μm (D–F, D′–F′); 1 μm (insets in D). (G–H) Montages of time lapse images at 1 SPF of (G) mGFP–Wsp1 and (H) Vrp1–mGFP patches splitting in bbc1Δ and bbc1Δ sla1Δ cells and not splitting in sla1Δ and WT cells. Scale bar: 1 μm. Arrowheads indicate instances of patch splitting. (I,J) Quantification of the percentage of events when (I) mGPF–Wsp1 or (J) Vrp1–mGFP patches internalized normally (blue), remained immobile (black), or split in two (red) in WT, bbc1Δ, sla1Δ and bbc1Δ sla1Δ cells. *P<0.001 for the difference from bbc1Δ (χ-squared test). (K) Diagram of proposed interactions of the Myo1 SH3 domain. (L,M) Effects of (L) vrp1Δ or bzz1Δ and (M) myo1ΔSH3-LCA (myo1Δ3CA) on the percentage of events when mGPF–Wsp1 patches internalized normally (blue), remained immobile (black) or split in two (red) in bbc1Δ sla1Δ cells. *P<0.001 for the difference from bbc1Δ sla1Δ (χ-squared test). N indicates number of patches analyzed.

We also observed infrequently in sla1Δ and, more often, in sla1Δ bbc1Δ cells elongated actin structures that we called actin plumes (Fig. 6D,D′; Movie 1). These are similar to enlarged actin structures found in S. cerevisiae sla1Δ bbc1Δ cells (Kaksonen et al., 2005). In addition to localizing to the base of these plumes, Myo1 associated with both the tip of the actin plumes and at the middle where the plume appeared to kink (Fig. 6D, inset). In single-color imaging of mGFP–Myo1 in sla1Δ and bbc1Δ sla1Δ cells, these plumes appeared as stable cytoplasmic dots of Myo1 swinging like a pendulum from stable Myo1 platforms on the membrane (Fig. 6E–F′; Movie 2). Thus, besides suppressing actin patch assembly, Sla1 appears to play a role in regulating Myo1 positioning at the endocytic site.

Wsp1 and Vrp1 remain with Myo1 at the base of invaginations in the absence of Bbc1

Close inspection of mGFP–Wsp1 dynamics revealed that this NPF also exhibits an unusual behavior but in bbc1Δ and bbc1Δ sla1Δ rather than in sla1Δ cells. Unlike S. cerevisiae Las17 and Vrp1 (Kaksonen et al., 2003, 2005), S. pombe Wsp1 and Vrp1 normally internalize with an endocytic vesicle for a few seconds before dissipating into the cytoplasm (Sirotkin et al., 2005). However, in bbc1Δ and bbc1Δ sla1Δ cells, Wsp1 and Vrp1 patches frequently split in two, with a fraction internalizing normally and another fraction remaining at the base of invagination (Fig. 6G,H; Fig. S6A, Movie 3). 84% and 95% of mGFP–Wsp1 patches split in bbc1Δ and bbc1Δ sla1Δ cells, respectively, while mGFP–Wsp1 patches rarely split in sla1Δ and wild-type cells (Fig. 6I). Vrp1–mGFP patches split at a similar frequency; 78% in bbc1Δ, 84% in bbc1Δ sla1Δ cells, but fewer than 15% of Vrp1–mGFP patches split in sla1Δ and wild-type cells (Fig. 6J).

Since Vrp1 can bind both Myo1 and Wsp1 and thereby bridge Myo1–Wsp1 interaction (Carnahan and Gould, 2003; Sirotkin et al., 2005), and Vrp1 and Bbc1 appear to compete for Myo1 binding (Fig. 4), splitting of Wsp1 patches in bbc1Δ cells could be due to enhanced Vrp1-mediated association of Wsp1 with Myo1 at the base of the endocytic invagination (Fig. 6K). To determine whether Vrp1 mediates Wsp1 patch splitting, we examined the effects of Vrp1 loss on Wsp1 patch splitting in bbc1Δ sla1Δ cells where Wsp1 patches are brighter and splitting is easier to observe (Figs 5D and 6G). Indeed, the frequency of Wsp1 patch splitting decreased from 92% in bbc1Δ sla1Δ cells to 21% in bbc1Δ sla1Δ vrp1Δ cells (Fig. 6L). In contrast, the loss of Bzz1, which is another Wsp1 binding partner that localizes to the base of endocytic invaginations (Arasada and Pollard, 2011; Idrissi et al., 2012; Kishimoto et al., 2011; Soulard et al., 2002; Sun et al., 2006), had no effect on Wsp1 patch splitting (Fig. 6L) or Wsp1 accumulation in patches in bbc1Δ sla1Δ cells (Fig. 5F). A reduction of Wsp1 patch splitting was also observed when vrp1Δ, but not bzz1Δ, was combined with just bbc1Δ, while in vrp1Δ alone cells, as in wild-type cells, Wsp1 patches rarely split (Fig. S6B).

To test whether the residual Wsp1 patch splitting observed in bbc1Δ sla1Δ vrp1Δ cells is due to direct binding of Wsp1 to the Myo1 SH3 domain that was observed by two-hybrid analysis (Carnahan and Gould, 2003), we combined bbc1Δ sla1Δ with the deletion of the Myo1 SH3 and LCA domains to disrupt the Myo1 interaction with both Vrp1 and Wsp1. As predicted, Wsp1 patch splitting completely ceased in bbc1Δ sla1Δ myo1ΔSH3-LCA cells (Fig. 6M), suggesting that both indirect Vrp1-mediated and direct binding of Wsp1 to Myo1 are responsible for Wsp1 patch splitting in the absence of Bbc1. In contrast, deletion of the CA domain resulted in only a minor effect on splitting Wsp1 patches, providing further evidence that Wsp1 patch splitting is mediated by Myo1 SH3 domain (Fig. S6C).

Endocytic structures internalize deeper in the absence of Bbc1

The observation that mGFP–Myo1 patches internalized nearly twice as far in bbc1Δ sla1Δ cells compared to sla1Δ cells (Fig. 6C) suggested that, similar to S. cerevisiae Bbc1 (Buser and Drubin, 2013; Kaksonen et al., 2005; Picco et al., 2018), S. pombe Bbc1 might have a role in regulating the length of the endocytic invagination. To determine the effect of bbc1Δ on the internalization of endocytic structures in S. pombe (Fig. 7A), we measured the accumulation of amphiphysin Hob1 and distances traveled by endocytic adaptors End4, Ent1 and Sla1 (Fig. 7; Fig. S7). Since Rvs167, an S. cerevisiae homolog of Hob1, binds around the tubular invagination (Idrissi et al., 2012, 2008; Kishimoto et al., 2011; Kukulski et al., 2012; Picco et al., 2015), accumulation of Hob1 provides a measure of the length of endocytic invagination. The three adaptors associate with the tip of invaginations (Boettner et al., 2011; Idrissi et al., 2012; Picco et al., 2015; Skruzny et al., 2012); therefore, the distance traveled by these markers represents the depth of endocytic internalizations. Compared to wild-type cells, Hob1–mGFP patch accumulation increased by 88% in bbc1Δ cells and 77% in bbc1Δ sla1Δ cells but it did not increase in sla1Δ cells (Fig. 7B,C). Additionally, the average distance traveled by Hob1–mGFP patches doubled in both bbc1Δ and bbc1Δ sla1Δ mutants compared to patches in wild-type and sla1Δ cells (Fig. 7B). Similarly, End4–mGFP and Ent1–mGFP patches internalized ∼2 times farther in bbc1Δ and bbc1Δ sla1Δ cells than in wild-type and sla1Δ cells: ∼1 µm in bbc1Δ and bbc1Δ sla1Δ cells versus ∼0.5 µm in wild-type and sla1Δ cells (Fig. 7D,E). The increased distance measured for End4–mGFP patches is not a result of the ability to track brighter patches longer since End4–mGFP patches had similar intensities and lifetimes in wild-type and bbc1Δ cells (Fig. S7A). Likewise, another invagination tip marker, Sla1, moved twice as far in bbc1Δ cells compared to wild-type cells (Fig. S7B). Collectively, these measurements are consistent with a 2-fold increase in the length of endocytic invaginations resulting in a 2-fold deeper internalizations of the endocytic structures in the absence of Bbc1. We hypothesized that this increase may be due to a prolonged association of Myo1 with Vrp1 and Wsp1, the main NPF at the endocytic site (Berro et al., 2010), at the base of endocytic invaginations. In agreement with this hypothesis, the distance traveled by the tip marker End4–mGFP returned to the normal 0.5 µm when bbc1Δ or bbc1Δ sla1Δ were combined with myo1ΔSH3-LCA (Fig. 7F), a mutant that disrupts Wsp1 retention at the base of invaginations (Fig. 6M). In contrast, End4–mGFP continued to internalize twice as far in bbc1Δ myo1ΔCA and bbc1Δ sla1Δ myo1ΔCA strains (Fig. 7G). Therefore, we conclude that the deeper internalizations observed in the absence of Bbc1 are mediated by the Myo1 SH3 domain, likely through the retention of Wsp1 and Vrp1 at the base of endocytic invaginations.

Fig. 7.

Bbc1 regulates the length of endocytic invaginations and distance traveled by endocytic structures. (A) A schematic of the proposed localization of Hob1, End4 and Ent1 at the endocytic site. (B) Average peak-aligned time courses of intensities and distances traveled for Hob1–mGFP in patches in WT (blue), bbc1Δ (red), sla1Δ (green) and bbc1Δ sla1Δ (purple) cells. (C) Bar graph showing average peak intensity of Hob1–mGFP in patches. (D,E) Distances traveled by (D) End4–mGFP and (E) Ent1–mGFP in endocytic patches in WT, bbc1Δ, sla1Δ and bbc1Δ sla1Δ cells. (F–G) Effect of (F) myo1Δ SH3-LCA (m1Δ3LCA) or (G) myo1ΔCA (m1ΔCA) on distance traveled by End4–mGFP in cells with different combinations of bbc1Δ and sla1Δ. Error bars represent s.d. N indicates number of patches analyzed. P-values in all panels represent statistical significance as determined by ANOVA with Tukey's post-hoc test. *P<0.0001, compared to WT.

Fig. 7.

Bbc1 regulates the length of endocytic invaginations and distance traveled by endocytic structures. (A) A schematic of the proposed localization of Hob1, End4 and Ent1 at the endocytic site. (B) Average peak-aligned time courses of intensities and distances traveled for Hob1–mGFP in patches in WT (blue), bbc1Δ (red), sla1Δ (green) and bbc1Δ sla1Δ (purple) cells. (C) Bar graph showing average peak intensity of Hob1–mGFP in patches. (D,E) Distances traveled by (D) End4–mGFP and (E) Ent1–mGFP in endocytic patches in WT, bbc1Δ, sla1Δ and bbc1Δ sla1Δ cells. (F–G) Effect of (F) myo1Δ SH3-LCA (m1Δ3LCA) or (G) myo1ΔCA (m1ΔCA) on distance traveled by End4–mGFP in cells with different combinations of bbc1Δ and sla1Δ. Error bars represent s.d. N indicates number of patches analyzed. P-values in all panels represent statistical significance as determined by ANOVA with Tukey's post-hoc test. *P<0.0001, compared to WT.

We initially identified the previously uncharacterized S. pombe protein Bbc1 in pulldowns of CR proteins. However, subsequent analysis revealed that Bbc1 does not localize to the CR, instead localizing to dynamic actin patches. While Bbc1 was originally discovered in S. cerevisiae as a myosin-1-tail-binding protein (Mochida et al., 2002), most of the subsequent work (Kaksonen et al., 2005; Picco et al., 2018) focused on the ability of Bbc1 to bind (Tong et al., 2002) and inhibit NPF activity of budding yeast WASp Las17 (Rodal et al., 2003). Thus, aside from mild inhibition of myosin-1 NPF activity (Sun et al., 2006), the significance of Bbc1 interaction with myosin-1 remained largely unknown. By taking advantage of the distinct behavior of Wsp1–Vrp1 and Myo1 in S. pombe, we provide evidence that the interaction of Bbc1 with myosin-1 is important for the role of Bbc1 as a negative regulator of endocytosis. Specifically, we propose a model (Fig. 8) where association of Bbc1 with Myo1 tail limits localization of Wsp1 and Vrp1 at the base of endocytic invagination, thereby preventing excessive tubulation of endocytic invaginations.

Fig. 8.

A model for regulation of actin patch assembly by Bbc1. (A) A graphical summary of the effects of bbc1Δ on the distribution and interactions of selected endocytic patch proteins and the endocytic site organization compared to WT. In WT cells, Bbc1 inhibits transient interaction of Wsp1 and Vrp1 with Myo1 at the base of invagination so that Wsp1 and Vrp1 internalize with the actin patch while Myo1 and Bbc1 remain at the base. In the absence of Bbc1 (bbc1Δ), interactions of Wsp1 and Vrp1 with Myo1 are enhanced, resulting in retention of a portion of Wsp1 and Vrp1 at the base of the invagination. This causes a denser branched actin network at the base of invagination, which provides the force generating longer endocytic invaginations. (B) A schematic diagram of Wsp1, Vrp1, Myo1 and Bbc1 protein domains and their known (solid lines) or proposed (dashed lines) binding interactions.

Fig. 8.

A model for regulation of actin patch assembly by Bbc1. (A) A graphical summary of the effects of bbc1Δ on the distribution and interactions of selected endocytic patch proteins and the endocytic site organization compared to WT. In WT cells, Bbc1 inhibits transient interaction of Wsp1 and Vrp1 with Myo1 at the base of invagination so that Wsp1 and Vrp1 internalize with the actin patch while Myo1 and Bbc1 remain at the base. In the absence of Bbc1 (bbc1Δ), interactions of Wsp1 and Vrp1 with Myo1 are enhanced, resulting in retention of a portion of Wsp1 and Vrp1 at the base of the invagination. This causes a denser branched actin network at the base of invagination, which provides the force generating longer endocytic invaginations. (B) A schematic diagram of Wsp1, Vrp1, Myo1 and Bbc1 protein domains and their known (solid lines) or proposed (dashed lines) binding interactions.

Bbc1 is part of an interconnected actin patch protein network

In S. pombe, Myo1 and Wsp1 are recruited to endocytic sites independently of one another but can interact with each other directly or indirectly via Vrp1. Vrp1 is recruited to patches by Wsp1 but also binds Myo1 (Carnahan and Gould, 2003; Sirotkin et al., 2005) and therefore can bridge Myo1–Wsp1 interaction. We hypothesize that these interactions of Myo1 with Wsp1 and Vrp1 take place before the initiation of endocytic internalization and help retain both Myo1 and Wsp1–Vrp1 in patches. Indeed, accumulation of Myo1 is reduced in the absence of Vrp1 and slightly increased in the absence of Bbc1, which also increases Vrp1 accumulation in patches. This interaction is likely transient since upon initiation of patch internalization, which occurs 2 s after Myo1, Wsp1 and Vrp1 reach peak assembly, Wsp1 and Vrp1 internalize with the patch, while Myo1 remains at the base of the invagination (Sirotkin et al., 2005, 2010).

Collectively, our results support a simple model based on competition of Bbc1 with Wsp1 and Vrp1 for the Myo1 tail (Fig. 8). Bbc1 completely depends on Myo1 for localization to patches, and specifically requires the Myo1 TH2 and SH3 domains. Bbc1, Vrp1, and Wsp1 but not other patch components associate with the Myo1 SH3–LCA fragment in our in vivo binding assay. In the absence of Bbc1, Vrp1 shows increased accumulation in patches, and Bbc1 shows increased accumulation in patches in the absence of Vrp1. Conversely, overexpression of Bbc1 reduces the levels of Vrp1, Wsp1 and Myo1 in patches, presumably by disrupting the interactions of Myo1 with Vrp1 and Wsp1. Most significantly, in the absence of Bbc1, Vrp1 and Wsp1 show an unusual splitting behavior where a portion of Vrp1 and Wsp1 remains with Myo1 at the base of invagination. Since this splitting behavior was reduced or eliminated in the absence of Vrp1 or the Myo1 SH3 domain, respectively, the splitting of Wsp1 and Vrp1 patches is likely due to enhanced interaction of Vrp1 and Wsp1 with Myo1 in the absence of competition from Bbc1. Future biochemical work is needed to verify this competition model with purified proteins, which is hindered by the large size and proline-rich nature of Bbc1.

Enhanced accumulation of Bbc1 in the absence of Vrp1 and vice versa strongly support our proposed competition mechanism. However, measured stoichiometry and an observed 40% increase in Bbc1 in patches in vrp1Δ suggest that at peak only ∼70 out of ∼360 Myo1 molecules are available for binding Bbc1 and Vrp1; 50 are occupied by Bbc1 and 20 by Vrp1. Then, in the absence of Bbc1, Vrp1 can access an additional 50 sites, increasing peak accumulation of Vrp1 over the normal number of 130 molecules (Sirotkin et al., 2010), in line with the observed ∼50% increase. The access to Myo1 tail may be limited by binding of other ligands or by intramolecular folding. The evidence for such intramolecular folding has been provided for amoeboid (Hwang et al., 2007; Jontes et al., 1998; Lee et al., 1999), mammalian (Stöffler and Bähler, 1998) and yeast (Grötsch et al., 2010) long-tailed class 1 myosins.

Competition for the Myo1 SH3 domain may also explain an unusual internalization of a portion of Myo1 observed in sla1Δ. We hypothesize that this behavior can be due to Myo1 binding to a proline-rich ligand, such as Wsp1 normally bound by the Sla1 SH3 domains.

Comparison with budding yeast

In S. cerevisiae, Bbc1 and Sla1 both inhibit Las17 in vitro (Rodal et al., 2003) and bbc1Δ sla1Δ cells show enhanced actin patch assembly, sometimes resulting in the formation of actin comet tails (Kaksonen et al., 2005). In S. pombe, either bbc1Δ or sla1Δ alone had no or only minor effects on the extent of actin patch assembly but together resulted in a 2-fold increase in actin assembly, consistent with the proposed role of Bbc1 and Sla1 as NPF inhibitors. However, this increase of actin patch assembly in bbc1Δ sla1Δ cells was accompanied by ∼2–3-fold increased accumulation of all NPFs, raising the possibility that the enhanced actin assembly may be the result of a greater concentration of NPFs in patches rather than loss of inhibition of NPF activity. Alternatively, enhanced patch accumulation of Myo1 and Wsp1 in bbc1Δ sla1Δ cells can be explained by a hypothetical positive-feedback loop linking actin assembly and NPF accumulation.

Unlike S. pombe Wsp1, S. cerevisiae WASp Las17 remains at the base of the endocytic invagination and does not internalize with the patch (Kaksonen et al., 2003, 2005; Picco et al., 2015). However, in S. cerevisiae bbc1Δ cells, as in S. pombe, patches of the WASp homolog Las17 split and endocytic vesicles internalize deeper (Kaksonen et al., 2005). While this behavior has been attributed to possible interactions between Las17 and coat proteins in the absence of Bbc1 (Kaksonen et al., 2005), Las17 splitting can also be explained by competition of Wsp1 and Bbc1 for the myosin-1 tail, similar to the mechanism we propose for S. pombe Wsp1. A recent super-resolution microscopy study (Mund et al., 2018) revealed that Las17 localizes in a donut-like pattern surrounding the endocytic coat and, in bbc1Δ cells, spreads to a wider area normally occupied by the myosin-1 Myo5 and Bbc1, suggesting increased association of Las17 with Myo5 in the absence of Bbc1. This, coupled with a larger membrane surface area internalized in bbc1Δ cells (Picco et al., 2018), suggests that the internalizing portion of splitting Las17 patches may represent increased internalization of the Las17-coated membrane area that is not normally internalized, while the non-internalizing portion represents Las17 associated with Myo5 bound at the wider ring.

S. cerevisiae Bbc1 was also shown to recruit the Tda2–Aim21 complex, which regulates capping protein recruitment (Farrell et al., 2017; Shin et al., 2018). However, this role of Bbc1 has not yet been explored in S. pombe.

Models of force generation at endocytic sites

In S. pombe, as in other organisms, actin polymerization provides force for overcoming membrane tension during endocytosis (Basu et al., 2014). In wild-type cells, prior to initiation of an endocytic invagination, Wsp1 and Myo1 colocalize at endocytic patches where they interact directly and indirectly via Vrp1 (Carnahan and Gould, 2003; Sirotkin et al., 2005). Vrp1 activates Myo1 NPF activity (Sirotkin et al., 2005), while Bzz1 stimulates Wsp1 NPF activity (Arasada and Pollard, 2011). As the invagination forms, Wsp1 and Vrp1 internalize, while Myo1 and Bzz1 remain on the membrane at the base of the invagination (Sirotkin et al., 2005). One model of force generation suggests that this separation results in two separate zones of branched actin assembly that push off of each other (Arasada and Pollard, 2011; Arasada et al., 2018).

An alternative model proposes that most of the force is generated in one zone at the base of the emerging invagination (Picco et al., 2015). Several lines of evidence favor this latter model. First, Wsp1 is a much stronger NPF than Myo1, which needs stimulation by Vrp1 (Sirotkin et al., 2005). Second, Wsp1 reaches peak accumulation 2 s before the onset of invagination, when Wsp1 and Myo1 still colocalize in the same zone (Sirotkin et al., 2010). Further, movement of Wsp1–Vrp1 with the patch would separate Wsp1 and Myo1 from their respective activators, Bzz1 and Vrp1.

In addition to splitting of Wsp1 and Vrp1 patches, endocytic patch proteins internalize twice as far in bbc1Δ cells. We hypothesize that the prolonged localization of Wsp1 and Vrp1 at the base of the invagination leads to continued stimulation of Wsp1 by Bzz1, further stimulation of Myo1 NPF activity by Vrp1, and orientation of the growing actin filament ends towards the plasma membrane and near Myo1, where Myo1 can exert an additional force driving membrane invagination. We propose that this membrane-directed actin assembly is ultimately responsible for the increased distance traveled by endocytic structures observed in bbc1Δ cells either through elongated invaginations or increased propulsion of endocytic vesicles away from the membrane.

In summary, we have provided evidence that the SH3 domains of endocytic proteins compete for their ligands and this competition plays an important role in regulating actin assembly. These observations underscore the importance of future work towards dissecting the complex network of SH3 domain interactions at endocytic sites (Fernandez-Ballester et al., 2009; Tong et al., 2002; Tonikian et al., 2009) and the role of these interactions in orchestrating the precise assembly of proteins driving endocytic internalization.

Yeast strain construction

S. pombe strains used in this study are listed in Table S2. LifeAct–mCherry was originally described in Huang et al. (2012). All other yeast strains were constructed by PCR-based gene tagging and genetic crosses. Deletions or fluorescent protein tags marked with selectable markers were introduced into endogenous chromosomal loci by homologous recombination of PCR-amplified gene tagging modules (Bähler et al., 1998; Wach et al., 1994). Amplified modules were transformed into cells by lithium acetate method (Keeney and Boeke, 1994) and successful integrants were isolated based on selectable markers and confirmed by PCR and sequencing.

To tag proteins at the C-termini with fluorescent protein tags, the stop codons of the targeted genes were replaced with gene tagging cassettes PCR-amplified from pFA6a-mGFP-kanMX6 (pJQW 85-4) for monomeric (m)GFP (Sirotkin et al., 2010; Wu et al., 2003), pFA6a-mCherry-kanMX6 (pKS390) or pFA6a-mCherry-natMX6 (pKS391) for mCherry (Snaith et al., 2005), and pFA6a-mNeonGreen-kanMX6 for mNeonGreen (Shaner et al., 2013; Willet et al., 2015). All C-terminally tagged proteins were expressed from endogenous loci under control of the native promoters.

Gene deletions were constructed by replacing gene open reading frames with the ura4+ cassette amplified from KS-ura4 (Bähler et al., 1998) for bbc1Δ::ura4+ and vrp1Δ::ura4+, kanMX6 cassette amplified from pFA6a-kanMX6 (Bähler et al., 1998) for bbc1Δ::kanMX6 and bzz1Δ::kanMX6, and natMX6 cassette amplified from pCR2.1-nat (Sato et al., 2005) for sla1Δ::natMX6.

The C-terminal truncations of Myo1 were constructed by replacing sequences encoding domains targeted for deletion with a stop codon followed by the ura4+ cassette amplified from KS-ura4 (Bähler et al., 1998). The unmarked myo1ΔTH2 allele was constructed by replacing the ura4+ cassette of myo1ΔTH2-SH3-LCA allele with the SH3-LCA-stop codon sequence amplified by PCR with primers flanked by 70 nt regions of homology to the end of TH1 domain and myo1+ 3′ UTR. The myo1ΔTH2 transformants were isolated by counterselection on EMM plates containing 2 mg/ml 5-fluoroorotic Acid (5-FOA) (Stark et al., 2013). The Myo1 domain boundaries were defined as previously described (Lee et al., 2000; Sirotkin et al., 2005): TH2, amino acids (aa) 967–1112; SH3, aa 1113–1163; L, aa 1164–1186; and CA, aa 1187–1217.

A gene expressing N-terminally tagged mCherry–Cdc15 from the native locus under control of endogenous Pcdc15 promoter was constructed by replacing the ura4+ cassette in a cdc15+/cdc15Δ::ura4+ diploid strain with the cassette consisting of 500 bp of the 5′ cdc15+ flanking region, the mCherry coding sequence, a GGGGSGGGGSG linker sequence, the cdc15+ coding sequence, and 500 bp of the 3′ cdc15+ flanking region. Haploid mCherry–Cdc15 integrants were isolated by 5-FOA selection and confirmed by sequencing.

Strains combining two or more deletion or tagged alleles were constructed by standard genetic crosses on malt extract (ME) plates and tetrad dissections on YES plates followed by screening for wanted gene combinations by replica plating onto appropriate selective plates, microscopy and PCR diagnostics. In some cases, myo1Δ and wsp1Δ cells were transformed with pUR-myo1+ and pUR-wsp1+ plasmids (Lee et al., 2000), respectively, which facilitate mating and are subsequently lost upon sporulation.

Plasmid construction

The Ppc89-Linker-mCherry cassette flanked by XhoI and NheI sites in pCR-BluntII-TOPO vector (Invitrogen) was created by In-Fusion cloning using ClonTech In-Fusion HD Cloning Kit (Takara Bio, USA) following the manufacturer's instructions. Briefly, a linear fragment encoding a start codon (bold), aa 261–783 of Ppc89, and a 5 aa GSGSG linker (italicized) was PCR-amplified from pREP81-ppc89 f5/r3 with the primers Ppc89F (5′-CCTTCACTCGAGATGACAATAGATGAAAACGGTAATGC-3′) and Ppc89R (5′-CTCGCCCTTGCTCACACCAGAACCAGAACCGCTAAAATCCTTACCAACGTTG-3′). The resulting Ppc89-Linker product was fused by recombination via the 15 nt homology regions (underlined in primer sequences) to the N-terminal sequence of previously cloned mCherry in pCR-BluntII-TOPO vector that was linearized by PCR amplification with the TmChF primer (5′-GTGAGGAAGGGGGAGGAG-3′) corresponding to mCherry aa 2-7 and TmChR primer (5′-CATCTCGAGTGAAGGGCGA-3′) corresponding to the start codon, XhoI site, and pCR-BluntII-TOPO MCS. The resulting Ppc89-Linker-mCherry fragment flanked by XhoI and NheI sites was cleaved from pCR-BluntII-TOPO vector and cloned in place of mGFP in the previously made pSGP573-3xPnmt1-mGFP-L-23A and -3A constructs (Siam et al., 2004) digested with XhoI and NheI. In the resulting constructs, sequences encoding the Myo1 TH2–SH3–LCA (23A) fragment (aa 967–1217) or the SH3-LCA (3A) fragment (aa 113–1217) are fused to the C-terminus of Ppc89-Linker-mCherry via a 7-aa linker with the sequence GASGTGS, and the expression is controlled by the thiamine-repressible 3×Pnmt1 promoter (Maundrell, 1990).

To make Ppc89-mCherry-LCA construct, the pSGP573-3×Pnmt1-Ppc89-L-mCherry-L-23A plasmid was linearized by PCR amplification with primers flanking and thereby removing the TH2–SH3–LCA sequence: the mChGSr primer (5′-GGATCCAGTACCGCTAGCA-3′) and SPBNot1d primer (5′-TGAGCGGCCGCTCTAGGT-3′). The Myo1 LCA sequence was amplified from genomic DNA with primers SPBm1LCAd (5′-AGCGGTACTGGATCCAAAGGTTCTACACCTCAAACCACC-3′) and SPBm1LCAr (5′-TAGAGCGGCCGCTCACCAATCTTCTTCTTCATCACT-3′). The Myo1 LCA sequence was fused in place of TH2–SH3–LCA sequence in linearized pSGP573-3xPnmt1-Ppc89-L-mCherry-L-23A plasmid by recombination via the 15 nt homology regions (underlined in primer sequence) using the In-Fusion HD Cloning Kit (Takara Bio).

Untagged Bbc1 overexpression constructs were made by subcloning the bbc1+ ORF into the XhoI and XmaI sites of the pREP3x vector (Forsburg, 1993). To N-terminally tag Bbc1 with mEGFP, the Bbc1 overexpression construct was linearized with XhoI and a fragment encoding mEGFP followed by a GGGGSGGGGSG linker fused in through Gibson cloning (Gibson et al., 2009).

All plasmid constructs were verified by sequencing and then electroporated into cells followed by selection on appropriate EMM plates containing 5 µg/ml thiamine and lacking uracil for pSGP573 constructs or leucine for pREP3x constructs.

Growth assays

Cells were grown to log phase at 25°C in YES medium. 4×107 cells were resuspended in 1 ml YES and then serially diluted 10-fold four times. Then, 2 µl of each dilution were spotted onto YES plates and, for drug sensitivity assays, YES plates containing 0.25 µM latrunculin A (Cayman Chemical) or 10 µg/ml benomyl (Sigma). Plates were incubated at different temperatures as indicated on Fig. S1.

Microscopy

Except for Figs 1A–D, 2B,C and Fig. S2C, imaging was performed on a UltraView VoX Spinning Disc Confocal System (PerkinElmer) mounted on a Nikon Eclipse Ti-E microscope with a 100×/1.4 NA PlanApo objective, equipped with a Hamamatsu C9100-50 EMCCD camera, and controlled by Volocity (PerkinElmer) software. Stably integrated S. pombe strains were grown to exponential phase [optical density at 595 nm (OD595)=0.2–1.0] in liquid YES medium (Sunrise) while shaking in the dark at 25°C over 2 days. Cells containing protein expression plasmids with the 3×Pnmt1 promoter were grown overnight at 25°C in liquid EMM medium (Sunrise) lacking uracil (for pSGP573 constructs) or leucine (for pREP3x constructs) and containing 5 µg/ml thiamine to suppress premature protein overexpression, washed next day three times with EMM lacking thiamine, and grown overnight at 25°C for 15–20 h in the absence of thiamine to induce protein expression in liquid EMM medium lacking uracil (for pSGP573 constructs) or leucine (for pREP3x constructs). For microscopy, cells from 0.5–1 ml of culture were collected by a brief 1-min centrifugation in a microfuge (2,000 g), washed with EMM, pelleted, and 5 µl of partly re-suspended pellet was placed onto a pad of 25% gelatin in EMM on a glass slide, covered by a coverslip and sealed with 1:1:1 vaseline:lanolin:paraffin (VALAP). Samples were imaged after a 5-min incubation to allow for partial depolarization of actin patches. For time-lapse imaging, single-color images were taken every second for 60–120 s and two-color images were taken every 2 s for 60–120 s. Z-series of images spanning the entire cell width were captured at 0.4 μm intervals.

Images in Fig. 1A,B, Figs S1C and S2C were acquired using a personal DeltaVision microscope system (GE Healthcare), which includes an Olympus IX71 microscope, 60×/1.42 NA PlanApo and 100×/1.40 NA UPlanSApo objectives, fixed- and live-cell filter wheels, a Photometrics CoolSnap HQ2 camera, and softWoRx imaging software. Images were acquired at 25–29°C. Images are maximum-intensity projections of z-sections spaced at 0.5 µm for Figs 1B and 1 µm for Fig. S2C that were deconvolved with 10 iterations. For images in Fig. 1A and Fig. S2C, 1 ml of log-phase cells grown overnight in YES at 25°C in a shaking water bath were collected by centrifugation (400 g for 1–4 min), resuspended in ∼10 µl YES, and spotted directly on glass slide. Time-lapse imaging in Fig. 1B was performed using an ONIX microfluidics perfusion system (CellASIC). Cells were grown and collected as for Fig. 1A except that 4×107 cells were resuspended in 1 ml YES. Then, 50 µl of cell suspension was loaded into Y04C plates for 5 s at 8 psi, and YES liquid medium flowed into the chamber at 5 psi throughout imaging. Time-lapse images were obtained at 4-min intervals.

Images in Figs 1D, 2B,C were acquired with a spinning disk system, which includes a Zeiss Axiovert200 m microscope, Yokogawa CSU-22 scan head, 63×/1.46 NA PlanApochromat and 100×/1.40 NA PlanApo oil immersion objectives, and Hamamatsu ImageEM-X2 camera. For end-on imaging, cells grown to log-phase in EMM plus adenine, uracil and leucine, were loaded to vertical chambers in a 4% MAUL agarose pad and imaged every 2 min. To image patches in Fig. 2B,C, cells were grown to log-phase at 25°C in MAUL and then mounted onto a 2% MAUL agarose pad for imaging.

For visualization of nuclei and septa in Fig. S1C, cells were fixed with ice-cold 70% ethanol and incubated at 4°C for at least 30 min. After washing with potassium-free PBS, fixed cells were incubated in 1 mg/ml Methyl Blue (MB) for 30 min at room temperature. MB-stained cells were pelleted by centrifugation (400 g for 4 min), resuspended in PBS, and mixed with DAPI in 1:1 ratio immediately prior to imaging.

Image analysis

Image analysis was performed in ImageJ (National Institutes of Health). To make sure that different mutant backgrounds did not alter the expression levels of tagged proteins and that imaging conditions remained stable throughout an imaging session, for all strains within each dataset, we measured average background-subtracted whole-cell intensities, which correspond to the tagged protein expression levels. For each time series, we measured whole cell intensities of five cells, subtracted these values for either extracellular background or the intensities of untagged wild-type cells, and averaged the background-subtracted values at each time point. All strains within each dataset had statistically similar whole-cell intensities.

The intensities and positions of fluorescent protein-tagged markers in individual endocytic actin patches were manually tracked throughout lifetime of each patch using a circular region of interest (ROI) with a 10-pixel diameter. Mean fluorescence intensities of patches were subtracted for mean cytoplasmic intensities measured in cell areas away from patches, and distances traveled by patches from the original positions were calculated. Initiation of actin patch internalization was defined as the moment the patch shifted its position by at least 2 pixels (180 nm). Except for measurements of the number of molecules, time courses of background-subtracted intensities and distances from origin for individual patches were aligned to the time of peak intensity (time=0) and averaged at each time point. Where noted, fluorescence intensities for each condition were normalized to the average peak intensity in the wild-type (WT) cells.

For the measurements of the numbers of molecules, strains combing Fim1–mCherry and one of the mGFP-tagged patch markers were imaged in a single medial plane at the rate of 1 frame every 2 s. Whole-cell intensities and the intensities and positions of patches were measured as described above. For these measurements, the time courses of cytoplasmic background-subtracted intensities and distances from origin for individual patches were aligned to the time Fim1–mCherry patch started moving (time=0), corresponding to the initiation of patch internalization. The intensities and distance values were averaged at each time point and the fluorescence intensities were converted into the numbers of molecules using a calibration curve constructed based on the previously measured average peak numbers of molecules of End4, Pan1, Wsp1 and Myo1 in patches, reported in Fig. 7 in Sirotkin et al. (2010).

Colocalization between mGFP-tagged Bbc1 or Cdc15 and mCherry-tagged Fim1 was measured over lifetimes of Fim1 patches by visual inspection and confirmed by using the Plot Z function to detect the presence of mGFP and mCherry signals within a 10-pixel wide ROI; 30 patches were analyzed for each strain.

The percentage of patches that were splitting was measured by manually following the fates of at least 80 patches for each condition. Patch splitting events were identified visually and confirmed by measuring fluorescence intensities along line scans across patches (Fig. S6A).

Mass spectrometry

Mass spectrometry analysis was performed as previously described in Roberts-Galbraith et al. (2009, 2010).

Statistical analysis

All fluorescence intensity and distance values are displayed as mean±s.d. The data are from single or, where indicated, multiple imaging experiments where all strains were imaged under identical conditions. The number of cells and patches analyzed are indicated in figure legends. The statistical significance of the data for datasets of only two conditions was tested by Student's t-test in Microsoft Excel. The statistical significance for datasets of more than two conditions was tested by one-way ANOVA with Tukey's post-hoc test in KaleidaGraph (Synergy Software). Both tests were two-tailed and the significance values are reported in figures and figure legends. The statistical significance of the patch splitting data was tested through a χ-squared analysis in Microsoft Excel.

We thank Ms Keely Macmillan for help with making Myo1 C-terminal truncations, Ms Elisabeth Barone for help with making strains and tracking patches for counting molecules, and the anonymous reviewers for helpful suggestions.

Author contributions

Conceptualization: C.D.M., M.C.M., K.L.G., V.S.; Methodology: C.D.M., M.C.M., M.J., V.S.; Validation: C.D.M., V.S.; Formal analysis: C.D.M., M.C.M., R.C., V.S.; Investigation: C.D.M., M.C.M., R.C., M.J., V.S.; Resources: K.L.G., V.S.; Data curation: C.D.M.; Writing - original draft: C.D.M.; Writing - review & editing: C.D.M., M.C.M., K.L.G., V.S.; Supervision: V.S.; Project administration: K.L.G., V.S.; Funding acquisition: K.L.G., V.S.

Funding

This work was supported by Public Health Service awards from the National Institute of General Medical Sciences (NIGMS) T32 GM007347 to the Vanderbilt MSTP, F31 GM119252 to M.C.M., R01 GM101035 and R35 GM131799 to K.L.G., and American Heart Association (AHA) SDG 11SDG5470024 to V.S. Deposited in PMC for release after 12 months.

Aghamohammadzadeh
,
S.
and
Ayscough
,
K. R.
(
2009
).
Differential requirements for actin during yeast and mammalian endocytosis
.
Nat. Cell Biol.
11
,
1039
-
1042
.
Anderson
,
B. L.
,
Boldogh
,
I.
,
Evangelista
,
M.
,
Boone
,
C.
,
Greene
,
L. A.
and
Pon
,
L. A.
(
1998
).
The Src homology domain 3 (SH3) of a yeast type I myosin, Myo5p, binds to verprolin and is required for targeting to sites of actin polarization
.
J. Cell Biol.
141
,
1357
-
1370
.
Arasada
,
R.
and
Pollard
,
T. D.
(
2011
).
Distinct roles for F-BAR proteins Cdc15p and Bzz1p in actin polymerization at sites of endocytosis in fission yeast
.
Curr. Biol.
21
,
1450
-
1459
.
Arasada
,
R.
,
Sayyad
,
W. A.
,
Berro
,
J.
and
Pollard
,
T. D.
(
2018
).
High-speed superresolution imaging of the proteins in fission yeast clathrin-mediated endocytic actin patches
.
Mol. Biol. Cell
29
,
295
-
303
.
Bähler
,
J.
,
Wu
,
J.-Q.
,
Longtine
,
M. S.
,
Shah
,
N. G.
,
McKenzie
,
A.
, III
,
Steever
,
A. B.
,
Wach
,
A.
,
Philippsen
,
P.
and
Pringle
,
J. R.
(
1998
).
Heterologous modules for efficient and versatile PCR-based gene targeting in Schizosaccharomyces pombe
.
Yeast
14
,
943
-
951
.
Basu
,
R.
and
Chang
,
F.
(
2011
).
Characterization of dip1p reveals a switch in Arp2/3-dependent actin assembly for fission yeast endocytosis
.
Curr. Biol.
21
,
905
-
916
.
Basu
,
R.
,
Munteanu
,
E. L.
and
Chang
,
F.
(
2014
).
Role of turgor pressure in endocytosis in fission yeast
.
Mol. Biol. Cell
25
,
679
-
687
.
Berro
,
J.
and
Pollard
,
T. D.
(
2014
).
Local and global analysis of endocytic patch dynamics in fission yeast using a new “temporal superresolution” realignment method
.
Mol. Biol. Cell
25
,
3501
-
3514
.
Berro
,
J.
,
Sirotkin
,
V.
and
Pollard
,
T. D.
(
2010
).
Mathematical modeling of endocytic actin patch kinetics in fission yeast: disassembly requires release of actin filament fragments
.
Mol. Biol. Cell
21
,
2905
-
2915
.
Boettner
,
D. R.
,
Friesen
,
H.
,
Andrews
,
B.
and
Lemmon
,
S. K.
(
2011
).
Clathrin light chain directs endocytosis by influencing the binding of the yeast Hip1R homologue, Sla2, to F-actin
.
Mol. Biol. Cell
22
,
3699
-
3714
.
Boettner
,
D. R.
,
Chi
,
R. J.
and
Lemmon
,
S. K.
(
2012
).
Lessons from yeast for clathrin-mediated endocytosis
.
Nat. Cell Biol.
14
,
2
-
10
.
Boulant
,
S.
,
Kural
,
C.
,
Zeeh
,
J.-C.
,
Ubelmann
,
F.
and
Kirchhausen
,
T.
(
2011
).
Actin dynamics counteract membrane tension during clathrin-mediated endocytosis
.
Nat. Cell Biol.
13
,
1124
-
1131
.
Buser
,
C.
and
Drubin
,
D. G.
(
2013
).
Ultrastructural imaging of endocytic sites in Saccharomyces cerevisiae by transmission electron microscopy and immunolabeling
.
Microsc. Microanal.
19
,
381
-
392
.
Carnahan
,
R. H.
and
Gould
,
K. L.
(
2003
).
The PCH family protein, Cdc15p, recruits two F-actin nucleation pathways to coordinate cytokinetic actin ring formation in Schizosaccharomyces pombe
.
J. Cell Biol.
162
,
851
-
862
.
Chang
,
F.
,
Woollard
,
A.
and
Nurse
,
P.
(
1996
).
Isolation and characterization of fission yeast mutants defective in the assembly and placement of the contractile actin ring
.
J. Cell Sci.
109
,
131
-
142
.
Collins
,
A.
,
Warrington
,
A.
,
Taylor
,
K. A.
and
Svitkina
,
T.
(
2011
).
Structural organization of the actin cytoskeleton at sites of clathrin-mediated endocytosis
.
Curr. Biol.
21
,
1167
-
1175
.
Engqvist-Goldstein
,
E. Y.
and
Drubin
,
D. G.
(
2003
).
Actin assembly and endocytosis: from yeast to mammals
.
Annu. Rev. Cell Dev. Biol.
19
,
287
-
332
.
Farrell
,
K. B.
,
McDonald
,
S.
,
Lamb
,
A. K.
,
Worcester
,
C.
,
Peersen
,
O. B.
and
Di Pietro
,
S. M.
(
2017
).
Novel function of a dynein light chain in actin assembly during clathrin-mediated endocytosis
.
J. Cell Biol.
216
,
2565
-
2580
.
Fernandez-Ballester
,
G.
,
Beltrao
,
P.
,
Gonzalez
,
J. M.
,
Song
,
Y.-H.
,
Wilmanns
,
M.
,
Valencia
,
A.
and
Serrano
,
L.
(
2009
).
Structure-based prediction of the Saccharomyces cerevisiae SH3-ligand interactions
.
J. Mol. Biol.
388
,
902
-
916
.
Forsburg
,
S. L.
(
1993
).
Comparison of Schizosaccharomyces pombe expression systems
.
Nucleic Acids Res.
21
,
2955
-
2956
.
Galletta
,
B. J.
,
Chuang
,
D. Y.
and
Cooper
,
J. A.
(
2008
).
Distinct roles for Arp2/3 regulators in actin assembly and endocytosis
.
PLoS Biol.
6
,
e1
.
Gibson
,
D. G.
,
Young
,
L.
,
Chuang
,
R.-Y.
,
Venter
,
J. C.
,
Hutchison
,
C. A.
, III
and
Smith
,
H. O.
(
2009
).
Enzymatic assembly of DNA molecules up to several hundred kilobases
.
Nat. Methods
6
,
343
-
345
.
Goode
,
B. L.
,
Eskin
,
J. A.
and
Wendland
,
B.
(
2015
).
Actin and endocytosis in budding yeast
.
Genetics
199
,
315
-
358
.
Grötsch
,
H.
,
Giblin
,
J. P.
,
Idrissi
,
F.-Z.
,
Fernández-Golbano
,
I.-M.
,
Collette
,
J. R.
,
Newpher
,
T. M.
,
Robles
,
V.
,
Lemmon
,
S. K.
and
Geli
,
M.-I.
(
2010
).
Calmodulin dissociation regulates Myo5 recruitment and function at endocytic sites
.
EMBO J.
29
,
2899
-
2914
.
Higgs
,
H. N.
and
Pollard
,
T. D.
(
2001
).
Regulation of actin filament network formation through ARP2/3 complex: activation by a diverse array of proteins
.
Annu. Rev. Biochem.
70
,
649
-
676
.
Huang
,
J.
,
Huang
,
Y.
,
Yu
,
H.
,
Subramanian
,
D.
,
Padmanabhan
,
A.
,
Thadani
,
R.
,
Tao
,
Y.
,
Tang
,
X.
,
Wedlich-Soldner
,
R.
and
Balasubramanian
,
M. K.
(
2012
).
Nonmedially assembled F-actin cables incorporate into the actomyosin ring in fission yeast
.
J. Cell Biol.
199
,
831
-
847
.
Hwang
,
K.-J.
,
Mahmoodian
,
F.
,
Ferretti
,
J. A.
,
Korn
,
E. D.
and
Gruschus
,
J. M.
(
2007
).
Intramolecular interaction in the tail of Acanthamoeba myosin IC between the SH3 domain and a putative pleckstrin homology domain
.
Proc. Natl. Acad. Sci. USA
104
,
784
-
789
.
Idrissi
,
F.-Z.
,
Grötsch
,
H.
,
Fernández-Golbano
,
I. M.
,
Presciatto-Baschong
,
C.
,
Riezman
,
H.
and
Geli
,
M.-I.
(
2008
).
Distinct acto/myosin-I structures associate with endocytic profiles at the plasma membrane
.
J. Cell Biol.
180
,
1219
-
1232
.
Idrissi
,
F.-Z.
,
Blasco
,
A.
,
Espinal
,
A.
and
Geli
,
M. I.
(
2012
).
Ultrastructural dynamics of proteins involved in endocytic budding
.
Proc. Natl. Acad. Sci. USA
109
,
E2587
-
E2594
.
Jontes
,
J. D.
,
Ostap
,
E. M.
,
Pollard
,
T. D.
and
Milligan
,
R. A.
(
1998
).
Three-dimensional structure of Acanthamoeba castellanii myosin-IB (MIB) determined by cryoelectron microscopy of decorated actin filaments
.
J. Cell Biol.
141
,
155
-
162
.
Kaksonen
,
M.
and
Roux
,
A.
(
2018
).
Mechanisms of clathrin-mediated endocytosis
.
Nat. Rev. Mol. Cell Biol.
19
,
313
-
326
.
Kaksonen
,
M.
,
Sun
,
Y.
and
Drubin
,
D. G.
(
2003
).
A pathway for association of receptors, adaptors, and actin during endocytic internalization
.
Cell
115
,
475
-
487
.
Kaksonen
,
M.
,
Toret
,
C. P.
and
Drubin
,
D. G.
(
2005
).
A modular design for the clathrin- and actin-mediated endocytosis machinery
.
Cell
123
,
305
-
320
.
Kaksonen
,
M.
,
Toret
,
C. P.
and
Drubin
,
D. G.
(
2006
).
Harnessing actin dynamics for clathrin-mediated endocytosis
.
Nat. Rev. Mol. Cell Biol.
7
,
404
-
414
.
Keeney
,
J. B.
and
Boeke
,
J. D.
(
1994
).
Efficient targeted integration at leu1-32 and ura4-294 in Schizosaccharomyces pombe
.
Genetics
136
,
849
-
856
.
Kishimoto
,
T.
,
Sun
,
Y.
,
Buser
,
C.
,
Liu
,
J.
,
Michelot
,
A.
and
Drubin
,
D. G.
(
2011
).
Determinants of endocytic membrane geometry, stability, and scission
.
Proc. Natl. Acad. Sci. USA
108
,
E979
-
E988
.
Kovar
,
D. R.
,
Sirotkin
,
V.
and
Lord
,
M.
(
2011
).
Three's company: the fission yeast actin cytoskeleton
.
Trends Cell Biol.
21
,
177
-
187
.
Kukulski
,
W.
,
Schorb
,
M.
,
Kaksonen
,
M.
and
Briggs
,
J. A. G.
(
2012
).
Plasma membrane reshaping during endocytosis is revealed by time-resolved electron tomography
.
Cell
150
,
508
-
520
.
Lee
,
W.-L.
,
Ostap
,
E. M.
,
Zot
,
H. G.
and
Pollard
,
T. D.
(
1999
).
Organization and ligand binding properties of the tail of Acanthamoeba myosin-IA. Identification of an actin-binding site in the basic (tail homology-1) domain
.
J. Biol. Chem.
274
,
35159
-
35171
.
Lee
,
W.-L.
,
Bezanilla
,
M.
and
Pollard
,
T. D.
(
2000
).
Fission yeast myosin-I, Myo1p, stimulates actin assembly by Arp2/3 complex and shares functions with WASp
.
J. Cell Biol.
151
,
789
-
800
.
Maundrell
,
K.
(
1990
).
nmt1 of fission yeast. A highly transcribed gene completely repressed by thiamine
.
J. Biol. Chem.
265
,
10857
-
10864
.
McMahon
,
H. T.
and
Boucrot
,
E.
(
2011
).
Molecular mechanism and physiological functions of clathrin-mediated endocytosis
.
Nat. Rev. Mol. Cell Biol.
12
,
517
-
533
.
Michelot
,
A.
,
Costanzo
,
M.
,
Sarkeshik
,
A.
,
Boone
,
C.
,
Yates
,
J. R.
, III
and
Drubin
,
D. G.
(
2010
).
Reconstitution and protein composition analysis of endocytic actin patches
.
Curr. Biol.
20
,
1890
-
1899
.
Mochida
,
J.
,
Yamamoto
,
T.
,
Fujimura-Kamada
,
K.
and
Tanaka
,
K.
(
2002
).
The novel adaptor protein, Mti1p, and Vrp1p, a homolog of Wiskott-Aldrich syndrome protein-interacting protein (WIP), may antagonistically regulate type I myosins in Saccharomyces cerevisiae
.
Genetics
160
,
923
-
934
.
Mund
,
M.
,
van der Beek
,
J. A.
,
Deschamps
,
J.
,
Dmitrieff
,
S.
,
Hoess
,
P.
,
Monster
,
J. L.
,
Picco
,
A.
,
Nedelec
,
F.
,
Kaksonen
,
M.
and
Ries
,
J.
(
2018
).
Systematic nanoscale analysis of endocytosis links efficient vesicle formation to patterned actin nucleation
.
Cell
174
,
884
-
896.e17
.
Naqvi
,
S. N.
,
Zahn
,
R.
,
Mitchell
,
D. A.
,
Stevenson
,
B. J.
and
Munn
,
A. L.
(
1998
).
The WASp homologue Las17p functions with the WIP homologue End5p/verprolin and is essential for endocytosis in yeast
.
Curr. Biol.
8
,
959
-
962
.
Pelham
,
R. J.
, Jr.
and
Chang
,
F.
(
2001
).
Role of actin polymerization and actin cables in actin-patch movement in Schizosaccharomyces pombe
.
Nat. Cell Biol.
3
,
235
-
244
.
Picco
,
A.
,
Mund
,
M.
,
Ries
,
J.
,
Nédélec
,
F.
and
Kaksonen
,
M.
(
2015
).
Visualizing the functional architecture of the endocytic machinery
.
eLife
4
,
e04535
.
Picco
,
A.
,
Kukulski
,
W.
,
Manenschijn
,
H. E.
,
Specht
,
T.
,
Briggs
,
J. A. G.
and
Kaksonen
,
M.
(
2018
).
The contributions of the actin machinery to endocytic membrane bending and vesicle formation
.
Mol. Biol. Cell
29
,
1346
-
1358
.
Ramesh
,
N.
,
Anton
,
I. M.
,
Hartwig
,
J. H.
and
Geha
,
R. S.
(
1997
).
WIP, a protein associated with wiskott-aldrich syndrome protein, induces actin polymerization and redistribution in lymphoid cells
.
Proc. Natl. Acad. Sci. USA
94
,
14671
-
14676
.
Roberts-Galbraith
,
R. H.
,
Chen
,
J.-S.
,
Wang
,
J.
and
Gould
,
K. L.
(
2009
).
The SH3 domains of two PCH family members cooperate in assembly of the Schizosaccharomyces pombe contractile ring
.
J. Cell Biol.
184
,
113
-
127
.
Roberts-Galbraith
,
R. H.
,
Ohi
,
M. D.
,
Ballif
,
B. A.
,
Chen
,
J. S.
,
McLeod
,
I.
,
McDonald
,
W. H.
,
Gygi
,
S. P.
,
Yates
,
J. R.
, III
and
Gould
,
K. L.
(
2010
).
Dephosphorylation of F-BAR protein Cdc15 modulates its conformation and stimulates its scaffolding activity at the cell division site
.
Mol. Cell
39
,
86
-
99
.
Rodal
,
A. A.
,
Manning
,
A. L.
,
Goode
,
B. L.
and
Drubin
,
D. G.
(
2003
).
Negative regulation of yeast WASp by two SH3 domain-containing proteins
.
Curr. Biol.
13
,
1000
-
1008
.
Rodal
,
A. A.
,
Kozubowski
,
L.
,
Goode
,
B. L.
,
Drubin
,
D. G.
and
Hartwig
,
J. H.
(
2005
).
Actin and septin ultrastructures at the budding yeast cell cortex
.
Mol. Biol. Cell
16
,
372
-
384
.
Rosenberg
,
J. A.
,
Tomlin
,
G. C.
,
McDonald
,
W. H.
,
Snydsman
,
B. E.
,
Muller
,
E. G.
,
Yates
,
J. R.
, III
and
Gould
,
K. L.
(
2006
).
Ppc89 links multiple proteins, including the septation initiation network, to the core of the fission yeast spindle-pole body
.
Mol. Biol. Cell
17
,
3793
-
3805
.
Sato
,
M.
,
Dhut
,
S.
and
Toda
,
T.
(
2005
).
New drug-resistant cassettes for gene disruption and epitope tagging in Schizosaccharomyces pombe
.
Yeast
22
,
583
-
591
.
Shaner
,
N. C.
,
Lambert
,
G. G.
,
Chammas
,
A.
,
Ni
,
Y.
,
Cranfill
,
P. J.
,
Baird
,
M. A.
,
Sell
,
B. R.
,
Allen
,
J. R.
,
Day
,
R. N.
,
Israelsson
,
M.
, et al. 
(
2013
).
A bright monomeric green fluorescent protein derived from Branchiostoma lanceolatum
.
Nat. Methods
10
,
407
-
409
.
Shin
,
M.
,
van Leeuwen
,
J.
,
Boone
,
C.
and
Bretscher
,
A.
(
2018
).
Yeast Aim21/Tda2 both regulates free actin by reducing barbed end assembly and forms a complex with Cap1/Cap2 to balance actin assembly between patches and cables
.
Mol. Biol. Cell
29
,
923
-
936
.
Siam
,
R.
,
Dolan
,
W. P.
and
Forsburg
,
S. L.
(
2004
).
Choosing and using Schizosaccharomyces pombe plasmids
.
Methods
33
,
189
-
198
.
Sirotkin
,
V.
,
Beltzner
,
C. C.
,
Marchand
,
J.-B.
and
Pollard
,
T. D.
(
2005
).
Interactions of WASp, myosin-I, and verprolin with Arp2/3 complex during actin patch assembly in fission yeast
.
J. Cell Biol.
170
,
637
-
648
.
Sirotkin
,
V.
,
Berro
,
J.
,
Macmillan
,
K.
,
Zhao
,
L.
and
Pollard
,
T. D.
(
2010
).
Quantitative analysis of the mechanism of endocytic actin patch assembly and disassembly in fission yeast
.
Mol. Biol. Cell
21
,
2894
-
2904
.
Skau
,
C. T.
and
Kovar
,
D. R.
(
2010
).
Fimbrin and tropomyosin competition regulates endocytosis and cytokinesis kinetics in fission yeast
.
Curr. Biol.
20
,
1415
-
1422
.
Skruzny
,
M.
,
Brach
,
T.
,
Ciuffa
,
R.
,
Rybina
,
S.
,
Wachsmuth
,
M.
and
Kaksonen
,
M.
(
2012
).
Molecular basis for coupling the plasma membrane to the actin cytoskeleton during clathrin-mediated endocytosis
.
Proc. Natl. Acad. Sci. USA
109
,
E2533
-
E2542
.
Snaith
,
H. A.
,
Samejima
,
I.
and
Sawin
,
K. E.
(
2005
).
Multistep and multimode cortical anchoring of tea1p at cell tips in fission yeast
.
EMBO J.
24
,
3690
-
3699
.
Sohrmann
,
M.
,
Fankhauser
,
C.
,
Brodbeck
,
C.
and
Simanis
,
V.
(
1996
).
The dmf1/mid1 gene is essential for correct positioning of the division septum in fission yeast
.
Genes Dev.
10
,
2707
-
2719
.
Soulard
,
A.
,
Lechler
,
T.
,
Spiridonov
,
V.
,
Shevchenko
,
A.
,
Shevchenko
,
A.
,
Li
,
R.
and
Winsor
,
B.
(
2002
).
Saccharomyces cerevisiae Bzz1p is implicated with type I myosins in actin patch polarization and is able to recruit actin-polymerizing machinery in vitro
.
Mol. Biol. Cell
22
,
7889
-
7906
.
Stark
,
B. C.
,
James
,
M. L.
,
Pollard
,
L. W.
,
Sirotkin
,
V.
and
Lord
,
M.
(
2013
).
UCS protein Rng3p is essential for myosin-II motor activity during cytokinesis in fission yeast
.
PLoS ONE
8
,
e79593
.
Stöffler
,
H.-E.
and
Bähler
,
M.
(
1998
).
The ATPase activity of Myr3, a rat myosin I, is allosterically inhibited by its own tail domain and by Ca2+ binding to its light chain calmodulin
.
J. Biol. Chem.
273
,
14605
-
14611
.
Sun
,
Y.
,
Martin
,
A. C.
and
Drubin
,
D. G.
(
2006
).
Endocytic internalization in budding yeast requires coordinated actin nucleation and myosin motor activity
.
Dev. Cell
11
,
33
-
46
.
Tong
,
A. H. Y.
,
Drees
,
B.
,
Nardelli
,
G.
,
Bader
,
G. D.
,
Brannetti
,
B.
,
Castagnoli
,
L.
,
Evangelista
,
M.
,
Ferracuti
,
S.
,
Nelson
,
B.
,
Paoluzi
,
S.
, et al. 
(
2002
).
A combined experimental and computational strategy to define protein interaction networks for peptide recognition modules
.
Science
295
,
321
-
324
.
Tonikian
,
R.
,
Xin
,
X.
,
Toret
,
C. P.
,
Gfeller
,
D.
,
Landgraf
,
C.
,
Panni
,
S.
,
Paoluzi
,
S.
,
Castagnoli
,
L.
,
Currell
,
B.
,
Seshagiri
,
S.
, et al. 
(
2009
).
Bayesian modeling of the yeast SH3 domain interactome predicts spatiotemporal dynamics of endocytosis proteins
.
PLoS Biol.
7
,
e1000218
.
Wach
,
A.
,
Brachat
,
A.
,
Pöhlmann
,
R.
and
Philippsen
,
P.
(
1994
).
New heterologous modules for classical or PCR-based gene disruptions in Saccharomyces cerevisiae
.
Yeast
10
,
1793
-
1808
.
Wagner
,
A. R.
,
Luan
,
Q.
,
Liu
,
S.-L.
and
Nolen
,
B. J.
(
2013
).
Dip1 defines a class of Arp2/3 complex activators that function without preformed actin filaments
.
Curr. Biol.
23
,
1990
-
1998
.
Weinberg
,
J.
and
Drubin
,
D. G.
(
2012
).
Clathrin-mediated endocytosis in budding yeast
.
Trends Cell Biol.
22
,
1
-
13
.
Willet
,
A. H.
,
McDonald
,
N. A.
,
Bohnert
,
K. A.
,
Baird
,
M. A.
,
Allen
,
J. R.
,
Davidson
,
M. W.
and
Gould
,
K. L.
(
2015
).
The F-BAR Cdc15 promotes contractile ring formation through the direct recruitment of the formin Cdc12
.
J. Cell Biol.
208
,
391
-
399
.
Wu
,
J.-Q.
,
Kuhn
,
J. R.
,
Kovar
,
D. R.
and
Pollard
,
T. D.
(
2003
).
Spatial and temporal pathway for assembly and constriction of the contractile ring in fission yeast cytokinesis
.
Dev. Cell
5
,
723
-
734
.
Wu
,
J.-Q.
,
Sirotkin
,
V.
,
Kovar
,
D. R.
,
Lord
,
M.
,
Beltzner
,
C. C.
,
Kuhn
,
J. R.
and
Pollard
,
T. D.
(
2006
).
Assembly of the cytokinetic contractile ring from a broad band of nodes in fission yeast
.
J. Cell Biol.
174
,
391
-
402
.
Yakura
,
M.
,
Ozoe
,
F.
,
Ishida
,
H.
,
Nakagawa
,
T.
,
Tanaka
,
K.
,
Matsuda
,
H.
and
Kawamukai
,
M.
(
2006
).
zds1, a novel gene encoding an ortholog of Zds1 and Zds2, controls sexual differentiation, cell wall integrity and cell morphology in fission yeast
.
Genetics
172
,
811
-
825
.
Young
,
M. E.
,
Cooper
,
J. A.
and
Bridgman
,
P. C.
(
2004
).
Yeast actin patches are networks of branched actin filaments
.
J. Cell Biol.
166
,
629
-
635
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information