Skip to main content
Elsevier Sponsored Documents logoLink to Elsevier Sponsored Documents
. 2019 Jan 21;29(2):202–216.e7. doi: 10.1016/j.cub.2018.11.053

Filopodome Mapping Identifies p130Cas as a Mechanosensitive Regulator of Filopodia Stability

Guillaume Jacquemet 1,, Aki Stubb 1, Rafael Saup 1, Mitro Miihkinen 1, Elena Kremneva 2, Hellyeh Hamidi 1, Johanna Ivaska 1,3,4,∗∗
PMCID: PMC6345628  PMID: 30639111

Summary

Filopodia are adhesive cellular protrusions specialized in the detection of extracellular matrix (ECM)-derived cues. Although ECM engagement at focal adhesions is known to trigger the recruitment of hundreds of proteins (“adhesome”) to fine-tune cellular behavior, the components of the filopodia adhesions remain undefined. Here, we performed a structured-illumination-microscopy-based screen to map the localization of 80 target proteins, linked to cell adhesion and migration, within myosin-X-induced filopodia. We demonstrate preferential enrichment of several adhesion proteins to either filopodia tips, filopodia shafts, or shaft subdomains, suggesting divergent, spatially restricted functions for these proteins. Moreover, proteins with phosphoinositide (PI) binding sites are particularly enriched in filopodia. This, together with the strong localization of PI(3,4)P2 in filopodia tips, predicts critical roles for PIs in regulating filopodia ultra-structure and function. Our mapping further reveals that filopodia adhesions consist of a unique set of proteins, the filopodome, that are distinct from classical nascent adhesions, focal adhesions, and fibrillar adhesions. Using live imaging, we observe that filopodia adhesions can give rise to nascent adhesions, which, in turn, form focal adhesions. We demonstrate that p130Cas (BCAR1) is recruited to filopodia tips via its C-terminal Cas family homology domain (CCHD) and acts as a mechanosensitive regulator of filopodia stability. Finally, we demonstrate that our map based on myosin-X-induced filopodia can be translated to endogenous filopodia and fascin- and IRSp53-mediated filopodia.

Keywords: filopodia, adhesion, SIM, p130Cas, filopodome, filopodia tip adhesion, mechanotransduction, stiffness, integrin

Highlights

  • A super-resolution microscopy screen reveals filopodia core and accessory proteins

  • FAK and paxillin are accessory, and not core, components of filopodia tip adhesions

  • PI(3,4)P2 is the prominent phosphatidylinositol species enriched at filopodia tips

  • p130Cas is a mechanosensitive regulator of filopodia stability


Jacquemet et al. perform super-resolution imaging of filopodia and define a map of core and accessory components based on a pool of target proteins linked to cell adhesion. They show that filopodia adhesions are distinct from classical adhesions and nucleate nascent adhesions and are mechanosensitive, requiring phospho-p130Cas for stabilization.

Introduction

The ability of cells to migrate in vivo is necessary for many physiological processes, including embryonic development, tissue homeostasis, and wound healing. Cell migration is also implicated in distinct pathological conditions, such as inflammation and cancer metastasis. To migrate, cells interact with their environment, the extracellular matrix (ECM), via adhesion receptors, such as integrins, which provide a physical link between the ECM and the actin cytoskeleton [1]. Integrin function is controlled by a conformational switch between active and inactive states that determines ECM ligand interaction and subsequent receptor signaling [2]. Integrin activation can be triggered from within the cell by several mechanisms, including the Rap1-RIAM-talin pathway. In 2D, integrin-ligand engagement leads to the assembly of large signaling platforms, termed focal adhesions (FAs), which are composed of hundreds of proteins collectively termed the adhesome [3, 4]. FAs are highly dynamic and complex structures that develop from force-dependent maturation of nascent adhesions at the leading edge and which undergo integrin-specific centripetal translocation to form fibrillar adhesions. Importantly, FAs not only provide anchorage but also represent integrin heterodimer-ligand-specific [5] and/or ECM-ligand-specific signaling nodes with mechanosensing functions [6] and therefore constitute ideal signaling platforms for ECM recognition.

Cell motility through complex 3D microenvironments also requires efficient probing of the cell surroundings, including ECM and neighboring cells, via specialized sensory protrusions, such as filopodia [7]. Filopodia, finger-like actin-rich protrusions widely used by cells in vivo during normal processes (e.g., development, immune surveillance, and wound healing) and also during tumorigenesis and cancer cell dissemination [7, 8], are often the very first point of contact between a cell and its immediate surroundings. To this end, filopodia contain cell-surface receptors, such as integrins, cadherins, and growth factor receptors, that can interact with, and interpret, a wide variety of cues. In the context of ECM recognition, cues include ECM gradients [9], ECM topography [10], or ECM stiffness [11, 12]. Thus, filopodia contribute to haptotaxis [9] and possibly to durotaxis [13]. Filopodia are very dynamic structures that stabilize upon ECM tethering and formation of an adhesive structure, at their tips, termed filopodia adhesion. ECM attachment at filopodia is dependent on integrins, which are actively transported to filopodia tips by the motor protein myosin-X (MYO10) [14]. In addition, filopodia stabilization requires both talin-mediated integrin activation and integrin downstream signaling at the filopodium tip [15]. However, the composition of filopodia adhesions remains poorly defined.

The composition and architecture of FAs have been extensively studied using both microscopy and mass-spectrometric-based strategies [4, 16]. Importantly, the compilation of FA components, either by literature curation (Geiger adhesome) [3] or by proteomic approaches (consensus adhesome) [4], have led to significant advances in our understanding of adhesion-mediated processes. Considering that filopodia are significant structures in vivo that are also implicated in various biological processes and pathologies, such as angiogenesis and cancer progression [8], a more detailed analysis of proteins recruited to filopodia adhesions is likely to be fundamental to better understand filopodia functions. Filopodia are relatively small and labile structures (1- to 5-μm length and 50- to 200-nm width) and are therefore difficult to purify in a scale sufficient to perform mass spectrometry analyses. Here, to characterize the composition of filopodia tip adhesions, we used a targeted approach and mapped the localization of 80 proteins, implicated in cellular adhesion or protein-membrane lipid interactions, using structured illumination microscopy (SIM).

Results

Mapping Protein Localization in Filopodia Using SIM

To identify proteins that localize to filopodia, we performed a SIM-based screen, spanning 80 putative regulators of cellular adhesion or protein-membrane lipid association. Proteins of interest (POIs) (Data S1) included known filopodia components, actin regulators, established FA components (Geiger adhesome) [3], and adhesion proteins consistently identified in multiple mass spectrometry studies (consensus adhesome) [4]. We chose to visualize all the POIs as GFP-fusion proteins, as it was not feasible to generate, validate, and optimize antibodies against all endogenous POIs. Cells adhering to fibronectin and transiently co-expressing a GFP-tagged POI and red fluorescent protein (RFP)-MYO10 (to induce and visualize filopodia tips) were stained for F-actin and imaged using SIM (Figures 1A and 1B). SIM images of each POI are provided as Supplemental Information (Data S1 and S2). Strikingly, 15 of the 38 consensus adhesome proteins [4] imaged did not display clear accumulation in FAs (Figure 1C; Data S2), suggesting that they may contribute to other cell-ECM interfaces. To study POI localization and distribution along filopodia, line intensity profiles, manually drawn from filopodium tip to base (Figures 1A and 1B), were obtained for >200 filopodia per POI. Importantly, to evaluate POI distribution across multiple cells, the brightness and contrast of each image was automatically adjusted using the brightest cellular structure as the upper limit. Line intensity profiles were used to determine the percentage of filopodia positive for each POI (Figure 1C), filopodia length (Figure S1), and to create a map highlighting the distribution of the POIs within filopodia (Figure 2; see STAR Methods for details). The ImageJ macro and R scripts used to perform these quantifications are available as supplemental files (Data S2).

Figure 1.

Figure 1

Mapping Adhesion Proteins to Filopodia Using Structured Illumination Microscopy

(A–C) U2OS cells expressing a GFP/RFP-tagged protein of interest (POI) and GFP/RFP-MYO10 were plated on fibronectin for 2 hr, stained for F-actin, and imaged using structured illumination microscopy (SIM). POI distribution within filopodia (from tip to base) was assessed with line intensity profiles (n numbers can be found in Data S1).

(A) An example illustrating the distribution of GFP-TLN1 (talin-1), MYO10-mScarlet (myosin-X), and F-actin within filopodia. Blue square highlights the region of interest (ROI) that is magnified; scale bars: (main) 10 μm; (inset) 1 μm; yellow line was used to measure the TLN1, MYO10, and F-actin intensity profiles.

(B) Results of the line intensity profiles from (A).

(C) The percentage of filopodia positive for each POI as determined from line intensity profiles. Colored circles indicate POI inclusion in the Geiger adhesome [3] (red circle) and/or the consensus adhesome [4] (blue circle) and/or accumulation in focal adhesions (FAs) (this study, yellow circle); “core filopodia proteins” and “accessory filopodia proteins” are labeled.

See also Data S1 and S2.

Figure 2.

Figure 2

Generation of a Filopodia Map Using Correlative Imaging and Intensity Profile Averaging

Heatmap demonstrating the subcellular localization of each POI within filopodia based on >250 intensity profiles (measured as in Figure 1; n numbers can be found in Data S1). POIs are labeled using their official human gene name. To create this map, intensity profiles of each POI were binned (40 bins per filopodium for each intensity profile) and then averaged (see STAR Methods for more details) and displayed as a heatmap. POIs are labeled using their official human gene name; filopodium tip (defined by MYO10) and filopodium shaft are highlighted. See also Figure S1 and Data S1 and S2.

Protein Mapping Reveals Classes of Core and Accessory Filopodia Proteins

Multiple established filopodia-localizing proteins, including talin-1 (TLN1) [17], formin-like protein 3 (FMNL3) [18], lamellipodin (RAPH1) [19], vasodilator-stimulated phosphoprotein (VASP) [20], mDia2 (DIAPH3) [21], and fascin (FSCN1) [22] were clearly detected with high resolution in filopodia, validating our approach. Our comprehensive mapping revealed that the proteins imaged here could be organized into three categories according to detection frequency within filopodia. The first category is composed of POIs that are primarily absent from filopodia (Data S2; Figure 1C), and due to low detection rate, we consider these proteins not to be filopodia proteins. The second category of POI is detected in a high proportion of filopodia (60%–100%), indicating that these proteins are core filopodia proteins (Data S2; Figure 1C). The third category of POI is those reliably detected but present in only a small fraction of filopodia (10%–40%), suggesting that these proteins may be accessory filopodia proteins contributing to filopodia-specific functions or defining subsets of biologically distinct filopodia (Data S2; Figure 1C).

Phosphoinositide PI(3,4)P2 Is Enriched at Filopodia Tips

To uncover common elements among the core filopodia proteins identified here, a protein domain enrichment analysis was performed (Figure 3A). This analysis revealed that proteins containing the four-point-one, ezrin, radixin, moesin (FERM) (24%) and/or SRC homology 3 (SH3) (21%) and/or pleckstrin homology (PH)-like (34%) domains are enriched in filopodia (Figure 3A). A strong enrichment of proteins containing PH-like domains led us to speculate that the phosphoinositide (PI) composition of filopodia could be a key contributor to filopodia function.

Figure 3.

Figure 3

Mapping of Phosphoinositides within Filopodia

(A) Functional annotation analysis (protein domain enrichments) of the core filopodia proteins (detected in ≥60% of filopodia) was performed using the INTERPRO database integrated within the Database for Annotation, Visualization, and Integrated Discovery (DAVID) platform.

(B) Distribution of phosphoinositides (PIs) in filopodia. U2OS cells transiently expressing MYO10-mScarlet and GFP-tagged probes, binding with high affinity to a single PI species (PI species, probe used; PI(3)P, GFP-FYVE-PH; PI(4)P, GFP-P4M; PI(4,5)P2, GFP-PLC(δ1)-PH; PI(3,4,5)P3, GFP-BTK-PH; PI(3,4)P2, GFP-TAPP-PH), were plated on fibronectin for 2 hr, fixed, stained for F-actin, and imaged using SIM. Maximum intensity projections (MIPs) are displayed; scale bars: (main) 20 μm; (inset) 2 μm; yellow arrows highlight filopodia tips.

(C) The distribution of each probe within filopodia was then analyzed and displayed as described in Figures 1 and 2.

(D) U2OS cells expressing Lifeact-mTurquoise2, GFP-TAPP-PH, and MYO10-mScarlet were plated on fibronectin and imaged live using an Airyscan confocal microscope (1 picture every 9 s; Video S1). Scale bars: (main) 15 μm; (inset) 5 μm.

(E) RAT2 cells expressing GFP-TAPP-PH were plated on fibronectin for 2 hr, stained for F-actin, and imaged using SIM. A representative MIP is displayed; scale bars: (main) 20 μm; (inset) 2 μm; yellow arrows highlight filopodia tips.

See also Data S1 and S2.

To map the distribution of the various PI in filopodia, GFP-tagged probes with high affinity to a single PI species [23] were imaged using SIM and analyzed as described above (Figures 3B and 3C). PI(3)P (imaged using GFP-FYVE-PH) was mostly detected on vesicular structures within the cell body, but not in filopodia (Figures 3B and 3C). PI(4)P (imaged using GFP-P4M) localized at the plasma membrane and on intracellular vesicles but was only weakly detected within filopodia (Figures 3B and 3C). In contrast, PI(4,5)P2 (labeled with GFP-PLCγPH) and PI(3,4,5)P3 (labeled with GFP-BTK-PH) were both strongly detected within filopodia with a relatively homogeneous distribution (similar to the plasma membrane). Strikingly, PI(3,4)P2 (labeled using GFP-TAPP-PH) was also detected in filopodia, but in contrast to the other PI, PI(3,4)P2 was strongly enriched to filopodia tips. Importantly, PI(3,4)P2 accumulation at filopodia tips was confirmed in live cells (Figure 3D; Video S1) and in endogenous filopodia (Figure 3E). This accumulation of PI(3,4)P2 to filopodia tips is surprising and suggests that PI(3,4)P2 contributes to filopodia tip functions or organization. Future work will aim at identifying the role of PI(3,4)P2 in filopodia as well as the proteins that are responsible for its accumulation at filopodia tips.

Video S1. PI(3,4)P2 Dynamics in Filopodia, Related to Figure 3

U2OS cells expressing MYO10-mScarlet, GFP-TAPP-PH and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope. The yellow square highlights a ROI, which is magnified.

Download video file (4.7MB, mp4)

SIM Mapping Reveals Novel Filopodia Tip Proteins and Subdomains within Filopodia Shafts

To quantitatively analyze the preferential recruitment of the core filopodia proteins, identified here, to filopodia tips or shafts, an enrichment ratio was calculated (Figure 4). As expected, proteins known to compose the tip complex, such as MYO10 [14], VASP [20], DIAPH3 [21], FMNL3 [18], and RAPH1 [19], were strongly enriched to filopodia tips (Figure 4). Importantly, our analysis revealed that F-actin is low in the filopodium tip compared to the filopodium shaft. Conversely, the amount of plasma membrane was slightly enriched at filopodium tips compared to shafts (Figures 2 and 4). This is likely due to the formation of a bud at the end of filopodia. Therefore, the enrichment score of a POI was compared to the enrichment score obtained for the plasma membrane rather than the enrichment score of F-actin (Figure 4).

Figure 4.

Figure 4

Preferential Enrichment of Adhesion Proteins to Filopodium Shaft or Tip

The preferential recruitment of core filopodia proteins to filopodia tips or shafts was assessed by calculating an enrichment ratio (averaged intensity at filopodium tip versus shaft). Results are displayed as Tukey boxplots using a logarithmic scale, and the POIs are ordered as a function of the median enrichment score. Enrichment scores of F-actin and the plasma membrane (labeled by CAAX-GFP) are highlighted in orange. A median enrichment score displaying ≥two-fold change over that of the plasma membrane was considered to represent strong POI enrichment to either filopodia tips (≥two-fold increase, highlighted in green) or to filopodia shafts (≥two-fold decrease, highlighted in blue). Statistically significant enrichment scores are noted as either “increased in filopodia tips” or “increased in filopodia shafts.” See also Data S1 and S2.

Our mapping revealed several original filopodia tip proteins, such as p130Cas (BCAR1), tensin-4 (TNS4), and crk (CRK). Other proteins displaying preferential recruitment to filopodia tips over filopodia shafts include the integrin activity modulators TLN1 and talin-2 (TLN2), kindlin-2 (FERMT2), and ICAP-1 (ITGB1BP1; Figures 2 and 4). Proteins strongly enriched to filopodia shafts include predominantly actin-regulating and cross-linking proteins, such as eplins (LIMA1a and LIMA1b), alpha-actinin-1 (ACTN1), plastin (PLS3), and ezrin (EZR) (Figures 2 and 4). Interestingly, eps8 (EPS8) and IRSp53 (BAIAP2), two regulators of filopodia formation, which are also known to interact with each other [24], appear to be spatially segregated within established filopodia as BAIAP2 accumulates at filopodia tips while EPS8 is enriched to filopodia shafts (Figures 2 and 4). Filopodia shafts can be segmented into subdomains depending on the degree of penetration of certain proteins throughout the filopodium (from the base of the shaft toward the tip). For instance, ACTN1 labels only the base (first 20%) of the shaft and proteins, such as EZR or LIMA1, label the first 50%–60% portion of the shaft (Figure 2). The segregation of the filopodia shaft into subdomains could indicate that these segments are also functionally different.

Filopodia Adhesions Are Distinct from Other Adhesions

In migrating cells, adhesion sites are highly dynamic structures that undergo a well-defined force-dependent maturation sequence from nascent adhesions to fibrillar adhesions [25]. Our SIM-based mapping allowed us to identify several established integrin binders and integrin activity regulators (TLN1 and TLN2; FERMT1 and FERMT2; and ITGB1BP1) as core filopodia proteins, while other integrin binders and regulators, such as tensin-1 (TNS1), tensin-3 (TNS3), MDGI (FABP3), filamin-a (FLNA), and nischarin (NISCH), were absent from filopodia. In addition, components of the signaling modules BCAR1-CRK and the Ilk-pinch-parvin complex (ILK, LIMS1, and PARVA; IPP complex) were detected in filopodia with similar distributions (Figures 1 and 2). Several proteins accumulating in FAs were also detected in most filopodia, including VASP, ACTN1, testin (TES), LASP-1 (LASP1), and vinculin (VCL).

Filopodia are regions of low force in the cell [7], and therefore, it is not surprising that proteins associated with mature and/or fibrillar adhesions were not identified as core filopodia proteins. These proteins include the actin-binding tensin isoforms (TNS1–3) [26], PDLIM1/5/7 [27], TRIP6 [27], zyxin (ZYX) [26], and palladin (PALLD) [28] (Figures 1 and 2). In addition, the major non-muscle myosins mediating cellular contractility, non-muscle myosin heavy chain IIa (MYH9) and non-muscle myosin heavy chain IIb (MYH10), were not detected in filopodia. Importantly, multiple proteins associated with the low force-bearing nascent adhesions, including paxillin (PXN) [29], FAK (PTK2) [30], or arp3 (ACTR3) [31], were classified as filopodia accessory, not core, proteins, as they were detected in <40% of filopodia (Figures 1 and 2). Thus, our SIM mapping indicates that filopodia adhesions consist of a unique set of proteins, the filopodome, and are distinct from classical nascent adhesions, FAs and fibrillar adhesions.

Filopodia Adhesions Nucleate Nascent Adhesions

The absence of PXN or PTK2 in >60% of filopodia was unexpected, as there are documented interactions between these two proteins and a large number of the core filopodia proteins identified here (Figure S2A). In line with our mapping data using GFP-tagged proteins, we found that endogenous ILK and TLN1/2 localize to filopodia and PXN and PTK2 were observed only in a small percentage of filopodia (Figures S2B–S2E). When PXN was detected within filopodia, its localization varied between being detected at the tip and/or in the shaft (Figure S2E), and we hypothesized that the variable localization of PXN in filopodia could be linked to filopodia dynamics. Previous work has reported MYO10 localization in nascent adhesion prior to filopodia elongation [32], but we did not observe this phenomenon here. Live-cell imaging revealed that unstable filopodia are devoid of PXN (Figure 5A; Video S2), whereas, in stable filopodia, PXN is initially detected briefly at the tip, followed by localization to, and increased clustering in, filopodia shafts, possibly due to actin retrograde flow. Upon lamellipodia advancement, these clusters of PXN gave rise to FAs (Figure 5A; Video S2). These data suggest that stabilized filopodia adhesions can trigger the nucleation of nascent adhesions.

Figure 5.

Figure 5

Filopodia Tip Adhesions

(A) U2OS cells expressing Lifeact-mTurquoise2, PXN-mEmerald, and MYO10-mScarlet were plated on fibronectin and imaged live using an Airyscan confocal microscope (1 picture every 30 s; scale bar, 5 μm; Video S2). Images highlight time points of interest in a magnified area; yellow arrows highlight filopodia tips.

(B) U2OS cells expressing MYO10-mScarlet were plated on fibronectin for 2 hr; stained for F-actin, phospho-BCAR1 (Y410), and endogenous PXN; and imaged using SIM. A representative MIP is displayed. The blue and yellow squares highlight ROIs, which are magnified; scale bars: (main) 20 μm; (inset) 2 μm.

(C–E) Live-cell imaging of U2OS cells transiently expressing MYO10-mScarlet together with either Lifeact-mTurquoise2 and BCAR1-eGFP (C; Video S3), Lifeact-mTurquoise2 and TLN1-eGFP (D; Video S4), or ITGA5-GFP (E; Video S5). Cells were plated on fibronectin and imaged live using an Airyscan confocal microscope. Yellow arrows highlight filopodia tips; scale bars: 5 μm.

(F) U2OS cells expressing MYO10-mScarlet together with either GFP-BCAR1 full-length (FL), BCAR1 deletion constructs, or GFP-PTK2-FAT (PTK2FAT) were plated on fibronectin for 2 hr, stained for F-actin, and imaged using SIM. BCAR1ΔSH3, BCAR1 SH3 domain deletion; BCAR1ΔCCHD, BCAR1 CCHD domain deletion; BCAR1ΔCCHDΔSH3, BCAR1 SH3 domain and CCHD deletion; BCAR1CCHD, BCAR1 CCHD domain alone. Images highlight magnified areas of interest; scale bar: 2 μm; full fields of view are available in Figure S4. Distribution of each construct within filopodia was analyzed and displayed as described in Figures 1 and 2.

See also Figures S2, S3, and S4.

Video S2. Paxillin Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet, PXN-GFP and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope. The white square highlights a ROI, which is magnified.

Download video file (14.7MB, mp4)

BCAR1 Is a Novel Component of the Filopodia Tip Complex

Talin-mediated integrin activation and linkage to the actin cytoskeleton are required for filopodia stabilization and maturation into adhesions [15]. As adhesion maturation strongly correlates with increasing forces exerted on the substrate, this prompted us to review our core filopodia proteins for other potential force sensors. BCAR1 (p130Cas), one of our filopodia core components (Figure 1) strongly enriched in filopodia tips (Figure 4), has been implicated as a mechanosensitive protein that becomes phosphorylated by SRC in response to mechanical stretch [33]. We first validated our mapping data by staining endogenous BCAR1 and found that it indeed localizes to filopodia tips (Figure S3A). In addition, a phosphorylation-specific antibody indicated that endogenous BCAR1 is phosphorylated at filopodia tips, where it co-localizes with MYO10, but not with PXN (Figures 5B and S3B). Live-cell imaging also confirmed our mapping data, as BCAR1 was always observed at filopodia tips, regardless of filopodia stability (Figure 5C; Video S3), in a similar fashion to TLN1 (Figure 5D; Video S4), and ITGA5 distribution was relatively homogeneous (Figure 5E; Video S5). Altogether, our data indicate that BCAR1 is a constitutive component of the filopodia tip complex (Figures 1 and 5), and its phosphorylation status suggests that it may play a role in ECM sensing in filopodia adhesions.

Video S3. BCAR1 Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet, BCAR1-GFP and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope.

Download video file (4.3MB, mp4)
Video S4. TLN1 Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet, TLN1-GFP and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope.

Download video file (8.3MB, mp4)
Video S5. ITGA5 Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet and ITGA5-GFP were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope.

Download video file (6.5MB, mp4)

BCAR1 Is Recruited to Filopodia Tips via Its CCHD

BCAR1 localization to FAs has been suggested to be regulated via direct interaction with PXN or PTK2 [34, 35], both of which are absent from most filopodia. In addition, BCAR1 recruitment to filopodia tips is insensitive to inhibition of SRC, PTK2, or cellular contractility (Figure S3C). Together, this suggested that BCAR1 localization to filopodia tips occurs via a distinct mechanism to its FA targeting. BCAR1 is composed of five principal domains, including an N-terminal SH3 domain followed by a proline-rich region, a substrate domain, a serine-rich region, and a C-terminal Cas family homology domain (CCHD) [36]. We analyzed the subcellular location of BCAR1 constructs lacking the SH3 domain and/or the CCHD and, as previously described, observed that constructs lacking the SH3 domain or the CCHD accumulated poorly to FAs, and the construct lacking both domains was completely absent from FAs (Figure S4) [37]. Importantly, constructs lacking the CCHD failed to accumulate to filopodia tips, and the construct lacking the SH3 domain accumulated to filopodia tips to the same extent as the full-length wild-type construct (Figures 5F and S4). This indicates that BCAR1 CCHD, but not the SH3 domain, is required to target BCAR1 to filopodia tips. Importantly, BCAR1 CCHD alone is sufficient for filopodia tip localization, demonstrating that BCAR1 CCHD is both a FA-targeting domain and a filopodia-tip-complex-targeting domain (Figure 5F).

BCAR1 Contributes to Filopodia Stabilization and Stiffness Sensing

Previously, we revealed that integrin activation as well as integrin downstream signaling are required for efficient filopodia formation and stabilization [15]. In order to investigate the potential role of BCAR1 in these processes, we silenced BCAR1 expression using two independent small interfering RNAs (siRNAs) (Figure 6A). In U2OS and MDA-MB-231 cells, silencing of BCAR1 led to an increase in the number of MYO10 filopodia (Figures 6B and 6C), and overall filopodia length remained unaffected (Figure S5A). Importantly, the effect of BCAR1 silencing on filopodia numbers could be rescued by expressing murine GFP-BCAR1, but not by expressing GFP-BCAR1 lacking the CCHD (Figure 6D). These data indicate that BCAR1, unlike TLN1 [15], is not essential for filopodia formation. However, BCAR1 is critical for efficient filopodia stabilization, as BCAR1-silenced cells displayed a higher proportion of unstable filopodia and a lower proportion of stable filopodia compared with control cells (Figure 6E).

Figure 6.

Figure 6

BCAR1 Regulates Environment Sensing at Filopodia Tips

(A) Efficiency of siRNA-mediated (oligos nos. 5 and 6) silencing of BCAR1 in U2OS cells.

(B and C) BCAR1-silenced (oligos nos. 5 and 6) U2OS (B) and MDA-MB-231 (C) cells transiently expressing MYO10-GFP were plated on fibronectin for 2 hr, fixed, and the number of MYO10-positive filopodia per cell was quantified (n > 65 cells, three biological repeats; ∗∗∗p value < 5.4 × 10−6).

(D) BCAR1-silenced (oligo no. 6) U2OS cells transiently expressing MYO10-mScarlet together with GFP, GFP-BCAR1, or GFP-BCAR1ΔCCHD were plated on fibronectin for 2 hr, fixed, and the number of MYO10-positive filopodia per cell was quantified (n > 69 cells, three biological repeats; ∗∗∗p value < 1.1 × 10−5).

(E) BCAR1-silenced (oligo no. 6) U2OS cells transiently expressing MYO10-GFP were plated on fibronectin and imaged live using an Airyscan confocal microscope (1 picture every 5 s; scale bar, 20 μm). Representative images at different time points are shown. For each condition, MYO10-positive particles were automatically tracked, and MYO10 spot lifetime (calculated as a percentage of the total number of filopodia generated per cell) was plotted and displayed as Tukey boxplots (see STAR Methods for details; three biological repeats, more than 40 cells per condition, ∗∗∗p value < 8.78 × 10−5).

(F–H) U2OS cells expressing MYO10-mScarlet (F) or MYO10-GFP (G and H) were plated on fibronectin-coated polyacrylamide gels of defined stiffness (0.5 kPa, soft; 50 kPa, stiff) for 2 hr.

(F) Cells were stained for F-actin and phospho-BCAR1 (Y410) and imaged using a spinning disk confocal microscope. MIPs are displayed; yellow squares highlight ROI, which are magnified; yellow arrows highlight filopodia tips; scale bars: (main) 20 μm; (inset) 5 μm.

(G) Cells were stained for F-actin, imaged using an Airyscan confocal microscope, and the number of MYO10-positive filopodia per cell was quantified (n > 81 cells, three biological repeats; ∗∗∗p value < 5.5 × 10−20). MIPs are displayed in Figure S5C.

(H) Live-cell imaging on an Airyscan confocal microscope (scale bars: 20 μm). Representative images at different time points are shown. For each condition, MYO10 spot lifetime was analyzed as in (E) (three biological repeats, more than 34 cells per condition; ∗∗∗p value < 9.4 × 10−4).

See also Figure S5 and Data S1 and S2.

BCAR1 silencing did not appear to impair the ability of U2OS cells to generate traction forces (Figure S5B). As BCAR1 Y410 phosphorylation is increased upon mechanical stretch, a process that enables cells to sense the mechanical properties of the ECM [33], and as BCAR1 is phosphorylated at filopodia tips (Figure 5B), we speculated that BCAR1 phosphorylation could be one of the signals, used at filopodia tips, to sense stiffness. Using fibronectin-coated polyacrylamide gels of defined stiffness (0.5 kPa, soft; 50 kPa, stiff), we found that BCAR1 Y410 phosphorylation was significantly higher at filopodia tips when cells were plated on a stiffer substrate compared to a softer environment (Figure 6F). Interestingly, when plated on a soft substrate, cells formed a much higher number of MYO10 filopodia (Figures 6G and S5C), most of which were unstable (Figure 6H). These data mirror the results obtained when BCAR1 is downregulated and cells are plated on stiff substrate (Figures 6B-6E). Taken together, our data clearly demonstrate that BCAR1 is part of the filopodia tip mechanosensitive machinery, which attenuates initiation of new filopodia downstream of filopodia stabilization.

Taking the Filopodome beyond MYO10-Induced Filopodia

Here, we used MYO10-induced filopodia as a model to characterize the composition of filopodia tip adhesions (filopodome). Importantly, we could validate our key findings in endogenous filopodia, including the presence of the filopodome components RAPH1, BCAR1, VASP, and TLN1 (Figures 7A and S6A) and the absence of PXN (Figure S6A). BCAR1 Y410 phosphorylation could also be detected at the tips of endogenous filopodia in RAT2, MCF10DCIS.com and U2OS cells (Figures 7B–7D) as well as at the tip of filopodia induced by FSCN1 or BAIP2 (Figures 7E and 7F). Taken together, these data clearly demonstrate that our mapping data can be translated to other filopodia subtypes.

Figure 7.

Figure 7

ECM Sensing in Distinct Filopodia Types

(A) RAT2 cells expressing BCAR1-GFP were plated on fibronectin for 2 hr, and their endogenous filopodia were stained for F-actin and imaged using SIM. A representative MIP is displayed.

(B–D) Cells were plated on fibronectin (B, RAT2 cells, 2 hr; C, MCF10DCIS.com, 2 hr; D, U2OS, 20 min) fixed, stained for F-actin and phospho-BCAR1 (Y410), and endogenous filopodia imaged using SIM. Representative MIPs are displayed.

(E–G) U2OS cells expressing GFP-FSCN1 (E), GFP-BAIP2 (F), or GFP-FMNL3 (G; all known filopodia-inducing proteins) were plated on fibronectin for 2 hr, stained for F-actin and phospho-BCAR1 (Y410), and imaged using SIM. Representative MIPs are displayed.

(H) U2OS cells expressing GFP-FMNL3 and mCherry-TLN2 were plated on fibronectin for 2 hr, stained for F-actin, and imaged using SIM. A representative MIP is displayed.

(I) Live-cell imaging of U2OS cells transiently expressing Lifeact-mTurquoise2 and MYO10-GFP or Lifeact-mTurquoise2 and FMNL3-GFP (Video S6). Cells plated on fibronectin were imaged live using an Airyscan confocal microscope. Images display a time point of interest as well as color-coded time projections. Scale bars: 20 μm.

(J and K) U2OS cells expressing GFP-FMNL3 (J) or GFP-MYO10 (K) were plated on fibronectin for 2 hr, fixed and stained for F-actin together with inactive (clone 4b4) or active (clone 12G10) β1 integrin, and imaged using SIM. MIPs are displayed.

For all panels, the yellow squares highlight ROIs, which are magnified; yellow arrows highlight filopodia tips; scale bars: (main) 20 μm; (inset) 2 μm. See also Figure S6.

Interestingly, we could not detect BCAR1 Y410 phosphorylation or TLN2 at the tips of FMNL3-induced filopodia (Figures 7G and 7H), which prompted us to further analyze their composition and dynamics. Live-cell imaging revealed that FMNL3-induced filopodia hover over the substrate but fail to stabilize (Figure 7I; Video S6). In line with the lack of TLN2, the integrin pool within FMNL3-induced filopodia appears to be mostly inactive compared to the integrin activity levels in MYO10-induced filopodia (Figures 7J and 7K). Absent or reduced integrin activation in FMNL3-induced filopodia likely explains the failure of these filopodia to stabilize. These data clearly demonstrate the importance of integrin inside-out signaling in activating integrins at filopodia tips, and future work will aim at deciphering the contribution of the filopodome components in regulating integrin activity.

Video S6. Dynamics of MYO10- and FMNL3-Induced Filopodia, Related to Figure 7

U2OS cells expressing lifeact-mTurquoise2 and MYO10-GFP or lifeact-mTurquoise2 and FMNL3-GFP were plated on fibronectin and imaged live using an Airyscan confocal microscope.

Download video file (7MB, mp4)

Discussion

Filopodia are sensory protrusions specialized in probing the cellular environment, including neighboring cells, chemokines, and the ECM. Here, to gain insight into the composition and biological function of filopodia tip adhesions, we mapped the sub-filopodia localization of 80 proteins linked to cell adhesion and migration using MYO10-induced filopodia as a model system (Figures 1, 2, and 4). Additionally, we validated our data in both endogenous filopodia and in filopodia induced by other factors (Figure 7); however, our map is unlikely to be applicable to all types of filopodia or filopodia-like protrusions. For instance, we report that FMNL3-induced filopodia do not form adhesions at their tips and therefore are substantially different from MYO10-induced filopodia (Figure 7). In addition, it is likely that the composition of filopodia tip adhesions described here will vary in function of the cell types and/or conditions used. For instance, as demonstrated for the adhesome [38], the ECM (biochemical composition and/or stiffness) engaged at filopodia tips is likely to tune the composition of the filopodome.

Our mapping revealed an enrichment of proteins binding to PI in filopodia (Figure 3). Mapping of multiple PI species identified spatial segregation of these PI within filopodia. PI(4,5)P2 and PI(3,4,5)P3 display a homogeneous distribution, whereas PI(3,4)P2 exhibits strong enrichment to filopodia tips (Figure 3). PI(4,5)P2 is an activator of both CDC42 and N-WASP, two promotors of filopodia formation [39], and PI(4,5)P2-rich lipid bilayers induce the elongation of filopodia-like structures in vitro [40]. PI(3,4,5)P3 can also induce filopodia by activating MYO10, and preventing PI(3,4,5)P3 generation by inhibiting phosphoinositide 3-kinase is sufficient to block filopodia formation [41]. The filopodia-tip localization of PI(3,4)P2 is an original observation, and its role in filopodia remains to be investigated. PI(3,4)P2 was reported to promote stability and maturation of invadopodia via the recruitment of Tks5 [42, 43]. Enrichment of PI(3,4)P2 to filopodia tips is likely to promote the accumulation of specific proteins. In our study, we found that lamellipodin (RAPH1), a known PI(3,4)P2 binder [19], accumulates to filopodia tips (Figures 2 and 4). In vitro, PI(3,4)P2 inhibits PI(4,5)P2-mediated actin polymerization [44], and therefore, PI(3,4)P2 could also play a role in constraining filopodia extension. Future work will aim at identifying the role of PI(3,4)P2 in filopodia as well as the proteins that are responsible for its accumulation in filopodia tips.

Integrin function is controlled by a conformational switch between active and inactive states that dictate integrin-ECM ligand interaction [2]. Here, we report that integrin activation, from within the cell, at filopodia, is key for filopodia stabilization and ECM sensing (Figure 7). Our mapping revealed a specific subset of known integrin binders and activity modulators that are recruited to filopodia. These include the integrin activators talins and kindlins as well as the integrin inactivators ICAP-1 (ITGB1BP1) and moesin (MSN) (Figure 2), all of which bind to the β1 integrin cytoplasmic tail on overlapping sites [45]. Future challenges will be to establish how integrin functions, including transport and activation, are coordinated by multiple adaptors within filopodia and what is the sequence and hierarchy of protein binding to the short cytoplasmic domains of integrin β subunits.

Filopodia sense ECM topography and/or ECM stiffness [10, 11, 12]. We previously observed that filopodia stabilization precedes FA maturation and that this process directs cell migration [15]. Here, we observed that PXN-positive adhesions could form in filopodia shafts, which, upon lamellipodia advancement [46], lead to the formation of FAs (Figure 5). As filopodia are widely used by cells migrating in fibrillar matrices [47, 48], it is tempting to speculate that the formation of PXN-positive adhesions in the filopodia shaft could be a mechanism by which cells sense matrix alignment. In the context of stiffness sensing, filopodia tip proteins TLN1 and BCAR1 have both previously been reported to be mechanosensitive [36]. Forces exerted by individual filopodia typically range from 5 to 25 pN (with a maximum potential of up to 2 nN) and are principally mediated by actin retrograde flow [49, 50, 51] and, upon maturation, by myosin contractility [52]. Accordingly, neither MYH9 nor MYH10 were detected within filopodia (Figures 1 and 2). In the case of TLN1, forces above 5 pN (15 pN for further activation) can induce conformational changes that trigger a switch from talin-RIAM to talin-vinculin complexes, leading to FA stabilization [53]. In the case of BCAR1, forces induce the stretching of the BCAR1 substrate domain, leading to its phosphorylation by SRC [33]. BCAR1 phosphorylation is not mediated by actomyosin contractility but instead depends on an intact actin cytoskeleton [54]. Importantly, here, we demonstrate that BCAR1 phosphorylation at filopodia tips is stiffness sensitive (Figure 6F). Although the exact amount of force required to activate BCAR1 remains unknown, the BCAR1 substrate domain is disorganized, and it is likely that the weak forces mediated by actin retrograde flow alone (5 pN in magnitude) are sufficient to mediate activation [54]. Due to the low force requirement for BCAR1 activation compared to TLN1, it is likely that, at the filopodia tip, BCAR1 will be one of the first mechanosensitive proteins to be activated. BCAR1 activation may then lead to activation of Rap1 [55, 56], which would, in turn, promote talin-mediated integrin activation, adhesion re-enforcement, and filopodia stabilization. Importantly, BCAR1-dependent filopodia stabilization functions as a feedback inhibitor for initiation of new filopodia nucleation.

Altogether, we have revealed that filopodia adhesions consist of a unique set of proteins, the filopodome, that are distinct from classical nascent adhesions, FAs, and fibrillar adhesions. Our mapping will be a valuable resource for future studies aimed at unravelling the biological relevance of filopodia in different developmental and pathological conditions involving cell motility and cellular responsiveness to environmental cues.

STAR★Methods

Key Resources Table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies

Mouse anti-human active β1 integrin 12G10 (1:100 for IF) Abcam catalog number: ab30394; RRID:AB_775726
Mouse anti-human inactive β1 integrin monoclonal 4B4 (1:200 for IF) Beckman Coulter catalog number: 6603113; RRID:AB_10638675
Mouse monoclonal anti-p130Cas (BCAR1, 1:100 for IF, 1:1000 for WB) Santa Cruz Biotechnology catalog number: SC-20029; RRID:AB_628064
Mouse monoclonal anti-talin-1 (TLN1, clone 8d4, 1:100 for IF) Sigma catalog number: T3287; RRID:AB_477572
Mouse monoclonal anti-FAK (PTK2, Clone 77, 1:100 for IF) BD Biosciences catalog number: 610087; RRID:AB_397494
Mouse monoclonal anti-α-tubulin (clone 12G10, 1:1000 for WB) The Developmental Studies Hybridoma Bank catalog number: 12G10; AB_1157911
Mouse monoclonal anti-paxillin (PXN, Clone 349, 1:100 for IF) BD Biosciences catalog number: 610051; RRID:AB_397463
Rabbit polyclonal antibody raised against Phospho-p130Cas (BCAR1, Tyr410) Cell signaling catalog number: 4011; RRID:AB_2274823
Rabbit monoclonal antibody raised against human ILK (clone EPR1592; 1:100 for IF) Abcam catalog number: ab76468; RRID:AB_2126930

Chemicals, Peptides, and Recombinant Proteins

SiR-actin Cytoskeleton catalog number: CY-SC001
Bovine plasma fibronectin Merck catalog number: 341631
Dimethylsulphoxide (DMSO) Sigma-Aldrich catalog number: D2650
SRC inhibitor (PP2) Selleckchem catalog number: S7008
FAK (PTK2) inhibitor (PF-573228) Selleckchem catalog number: S2013
The myosin II inhibitor blebbistatin STEMCELL Technologies catalog number: 72402
Lipofectamine 3000 and the P3000TM Enhancer Reagent Thermo Fisher Scientific catalog number: L3000015
Human EGF Sigma-Aldrich catalog number: E9644
Hydrocortisone Sigma-Aldrich catalog number: H0888-1G
Cholera toxin Sigma-Aldrich catalog number: C8052-1MG
Insulin Sigma-Aldrich catalog number: I9278-5ML
Penicillin/streptomycin Sigma-Aldrich catalog number: P0781-100ML
Silane A-174 Sigma-Aldrich catalog number: 440159-100ML
Acrylamide solution Sigma-Aldrich catalog number: A4058
N, N′-Methylenebisacrylamide solution Sigma-Aldrich catalog number: M1533
TEMED Sigma-Aldrich catalog number: T9281
Sulfo-SANPAH Thermo Scientific catalog number: 22589
N-(3-Dimethylaminopropyl)-N’ ethylcarbodiimide hydrochloride (EDC) Sigma-Aldrich catalog number: 03450

Experimental Models: Cell Lines

U2OS osteosarcoma cells Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig DE catalog number: ACC 785
MDA-MB-231 triple-negative human breast adenocarcinoma ATCC catalog number: HTB-26
MCF10 DCIS.COM J.F. Marshall (Barts Cancer Institute, Queen Mary University of London, London, England, UK) [57] N/A
RAT2 cells (Rat embryonic fibroblasts) ATCC catalog number: CRL-1764

Oligonucleotides

The siRNA used as control (siCTRL) was Allstars negative control siRNA. QIAGEN catalog number: 1027281
siBCAR1#5 (Hs_BCAR1_5) QIAGEN catalog number: SI02757734
siBCAR1#6 (Hs_BCAR1_6) QIAGEN catalog number: SI02757741

Recombinant DNA

eGFP-TNS1 Previous study [58] N/A
eGFP-TNS2 Previous study [58] N/A
eGFP-TNS3 Previous study [59] N/A
eGFP-TNS4 Previous study [58] N/A
GFP-Sharpin Previous study [60] N/A
GFP-MDGI Previous study [61] N/A
GFP-FAK-FAT David D. Schlaepfer (UC San Diego Health, US) N/A
GFP-FL-FAK David D. Schlaepfer (UC San Diego Health, US) N/A
EFHD2-GFP Dirk Mielenz (University of Erlangen-Nuremberg, DE) [62] N/A
CAAX-GFP Gregory Giannone (Bordeaux University, FR) N/A
EGFP-Talin-1 Ben Goult (University of Kent, UK) N/A
mCherry-Talin-2 Ben Goult (University of Kent, UK) N/A
pDsRedC1-Kindlin-1 Ben Goult (University of Kent, UK) N/A
FMNL2-GFP Robert Grosse (University of Marburg, DE) [63] N/A
FMNL3-GFP Henry Higgs (Geisel School of Medicine at Dartmouth, US) [18] N/A
PPFIA1-GFP Guido Serini (University of Torino, IT) N/A
pEGFP-C1-Lamellipodin Matthias Krause (King’s College London, UK) [19] N/A
pEGFP-C2-Myo15a Jonathan Bird (NIH, Bethesda US) [64] N/A
GFP-ICAP-1 Daniel Bouvard (University of Grenoble, FR) N/A
GFP-KANK1 Reinhard Fässler (Max Planck Institute of Biochemistry, Martinsried, DE) [65] N/A
GFP-KANK2 Reinhard Fässler (Max Planck Institute of Biochemistry, Martinsried, DE) [65] N/A
GFP-KANK3 Reinhard Fässler (Max Planck Institute of Biochemistry, Martinsried, DE) [65] N/A
GFP-KANK4 Reinhard Fässler (Max Planck Institute of Biochemistry, Martinsried, DE) [65] N/A
BTK-PH-EGFP Matthias Wymann (University of Basel, Switzerland) N/A
PLC(δ1)-PH-EGFP Matthias Wymann (University of Basel, Switzerland) N/A
EGFP-tagged tandem FYVE Harald Stenmark (Oslo University Hospital) [66] N/A
GFP-Cas-wt Daniel Rösel (Charles University in Prague, Czech Republic) [37] N/A
GFP-CasdeltaCCH Daniel Rösel (Charles University in Prague, Czech Republic) [37] N/A
GFP-CasdeltaSH3 Daniel Rösel (Charles University in Prague, Czech Republic) [37] N/A
GFP-Cas-deltaCCH-deltaSH3 Daniel Rösel (Charles University in Prague, Czech Republic) [37] N/A
Kindlin-2-GFP Maddy Parsons (King’s College London, UK) N/A
Ezrin-GFP Maddy Parsons (King’s College London, UK) N/A
Vinculin-GFP Maddy Parsons (King’s College London, UK) N/A
Moesin-GFP Buzz Baum (University College London, UK) N/A
Lifeact-mTurquoise2 Joachim Goedhart (University of Amsterdam, NL) [67] N/A
Integrin alpha5-GFP Rick Horwitz (Allen institute for cell science, US) N/A
mEmerald-Alpha-Actinin-19 Addgene (Michael Davidson) catalog number: 53989
mEmerald-Fascin-C-10 Addgene (Michael Davidson) catalog number: 54094
pGFP Cas Addgene (Kenneth Yamada) catalog number: 50729
mEmerald-Cofilin-C-10 Addgene (Michael Davidson) catalog number: 54047
mEmerald-Coronin1B-C-10 Addgene (Michael Davidson) catalog number: 54049
pGFP CrkII Addgene (Kenneth Yamada) catalog number: 50730
mEmerald-Cortactin-C-12 Addgene (Michael Davidson) catalog number: 54051
mEmerald-mDia1-C-14 Addgene (Michael Davidson) catalog number: 54156
mEmerald-mDia2-C-14 Addgene (Michael Davidson) catalog number: 54158
mEmerald-Migfilin-C-14 Addgene (Michael Davidson) catalog number: 54181
pEGFP-IQGAP1 Addgene (David Sacks) [68] catalog number: 30112
mEmerald-LASP1-C-10 Addgene (Michael Davidson) catalog number: 54141
EGFP-EPLIN alpha Addgene (Elizabeth Luna) catalog number: 40947
EGFP-EPLIN beta Addgene (Elizabeth Luna) catalog number: 40948
mEmerald-PINCH-C-14 Addgene (Michael Davidson) catalog number: 54229
mEmerald-MyosinIIA-C-18 Addgene (Michael Davidson) [69] catalog number: 54190
mEmerald-MyosinIIB-C-18 Addgene (Michael Davidson) catalog number: 54192
mEmerald-Palladin-C-7 Addgene (Michael Davidson) catalog number: 54213
mEmerald-Parvin-C-14 Addgene (Michael Davidson) catalog number: 54214
GFP-PTEN Addgene (Alonzo Ross) [70] catalog number: 13039
mEmerald-Paxillin-22 Addgene (Michael Davidson) [71] catalog number: 54219
mEmerald-TES-C-14 Addgene (Michael Davidson) catalog number: 54276
mEmerald-VASP-N-10 Addgene (Michael Davidson) catalog number: 54297
mEmerald-Zyxin-6 Addgene (Michael Davidson) catalog number: 54319
E-cadherin-GFP Addgene (Jennifer Stow) [72] catalog number: 28009
pEGFP C1-Eps8 WT Addgene (Giorgio Scita) [73] catalog number: 74950
GFP-P4M-SidM Addgene (Tamas Balla) [74] catalog number: 51469
pcDNA3.1-6His-MyoX Addgene (Emanuel Strehler) [75] catalog number: 47607
ARP3-GFP Addgene (Matthew Welch) [76] catalog number: 8462
mScarlet-MYO10 This study (Data S1) N/A
pEGFP-CasCCHD This study (Data S1) N/A
mEmerald-MYO7A This study N/A
mEmerald-BAIAP2 This study N/A
mEmerald-PDLIM5 This study N/A
mEmerald-FERMT2 This study N/A
mEmerald-FHL2 This study N/A
mEmerald-FHL3 This study N/A
mEmerald-PDLIM7 This study N/A
mEmerald-PLS3 This study N/A
mEmerald-TRIP6 This study N/A
mEmerald-ALYREF This study N/A
mEmerald-ANXA1 This study N/A
mEmerald-BRIX1 This study N/A
mEmerald-DIMT1 This study N/A
mEmerald-FAU This study N/A
mEmerald-HP1BP3 This study N/A
mEmerald-PCOLCE This study N/A
mEmerald-POLDIP3 This study N/A
mEmerald-SERPINE1 This study N/A
mEmerald-SORBS1 This study N/A
mEmerald-TGM2 This study N/A
mEmerald-P4HB This study N/A
mEmerald-PDLIM1 This study N/A
mEmerald-SYNCRIP This study N/A
mEmerald-NISCH This study N/A
mEmerald-PRSS23 This study N/A

Software and Algorithms

Filopodia mapping This study (Data S2) N/A
RStudio (1.0.153) Foundation for Open Access Statistics. https://www.rstudio.com/
DAVID platform [77] https://david.ncifcrf.gov/
BoxPlotR [78] http://shiny.chemgrid.org/boxplotr/
PlotsOfData [79] https://huygens.science.uva.nl/PlotsOfData/
Fiji [80] https://fiji.sc/
Fiji plugin TrackMate [81] https://imagej.net/TrackMate
MATLAB (R2018b) MathWorks https://se.mathworks.com/products/matlab.html
TFM analysis (MATLAB software) Timo Betz (University of Münster) [82] N/A

Other

Glass-bottom dishes, High Tolerance 1.5 coverslip MatTek Corporation catalog number: P35G-0.170-14-C
Glass-bottom dishes, 1.0 coverslip MatTek Corporation catalog number: P35G-1.0-14-C
Polyacrylamide gels of defined stiffness; 0.5 kPa Matrigen catalog number: SV3510-EC-0.5
Polyacrylamide gels of defined stiffness; 50 kPa Matrigen catalog number: SV3510-EC-50
VECTASHIELD Vector laboratories catalog number: H-1000
FluoSpheres (505/515) Life Technologies catalog number: F881
Horse serum GIBCO BRL catalog number: 16050-122

Contact for Reagent and Resource Sharing

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Johanna Ivaska (johanna.ivaska@utu.fi).

Experimental Model and Subject Details

U2OS osteosarcoma cells, MDA-MB-231 (triple-negative human breast adenocarcinoma) cells and RAT2 cells (Rat embryonic fibroblasts) were grown in DMEM supplemented with 10% FCS. MCF10 DCIS.COM (DCIS.COM) cells were cultured in a 1:1 mix of DMEM (Sigma-Aldrich) and F12 (Sigma-Aldrich) supplemented with 5% horse serum (16050-122; GIBCO BRL), 20 ng/ml human EGF (E9644; Sigma-Aldrich), 0.5 mg/ml hydrocortisone (H0888-1G; Sigma-Aldrich), 100 ng/ml cholera toxin (C8052-1MG; Sigma-Aldrich), 10 μg/ml insulin (I9278-5ML; Sigma-Aldrich), and 1% (vol/vol) penicillin/streptomycin (P0781-100ML; Sigma-Aldrich). U2OS cells were purchased from DSMZ (Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig DE, ACC 785). MDA-MB-231 and RAT2 cells were provided by ATCC. DCIS.COM were provided by J.F. Marshall (Barts Cancer Institute, Queen Mary University of London, London, England, UK). All cells were tested for mycoplasma contamination. Cell lines have not been authenticated.

Method Details

Plasmids and transfection

Plasmids of interest were transfected using Lipofectamine 3000 and the P3000TM Enhancer Reagent (Thermo Fisher Scientific) according to the manufacturer’s instructions. The expression of proteins of interest was suppressed using 100 nM siRNA and lipofectamine 3000 (ThermoFisher Scientific) according to manufacturer’s instructions. The siRNA used as control (siCTRL) was Allstars negative control siRNA (QIAGEN, Cat. No. 1027281). The siRNAs targeting BCAR1 were purchased from QIAGEN (siBCAR1#5, Hs_BCAR1_5 FlexiTube siRNA, Cat. No. SI02757734; siBCAR1#6, Hs_BCAR1_6 FlexiTube siRNA, Cat. No. SI02757741).

Plasmids

The mScarlet-MYO10 construct was generated by inserting a gene block containing the mScarlet sequence (IDT, see Data S2 for sequence) [83] into pcDNA3.1-6His-MyoX using the KpnI restriction site. The BCAR1 CCHD construct was generated by inserting a gene block containing the BCAR1 CCHD sequence (IDT, see see Data S2 for sequence) into pEGFP-C1 using the XhoI and BamHI restriction sites. Several of the GFP tagged constructs, used here, were generated by the Genome Biology Unit core facility cloning service (Research Programs Unit, HiLIFE Helsinki Institute of Life Science, Faculty of Medicine, University of Helsinki, Biocenter Finland) by transferring entry clones from the ORFeome collaboration library into mEmerald destination vectors using the standard LR reaction protocol. Entry clone (I.M.A.G.E. Consortium CloneID [68],) transferred into pcDNA6.2/N-emGFP-DEST include MYO7A (100069043), BAIAP2 (100006086), PDLIM5 (100003316), FERMT2 (100067023), FHL2 (100006502), FHL3 (100003225), PDLIM7 (100004765), PLS3 (100003800), TRIP6 (100005523), ALYREF (100066457), ANXA1 (100003803), BRIX1 (100004738), DIMT1 (100004112), FAU (100005039), HP1BP3 (100067555), PCOLCE (100004553), POLDIP3 (100000169), SERPINE1 (100003324) and SORBS1 (100064166). Entry clones transferred into pcDNA6.2/C-emGFP-DEST include TGM2 (100074142), P4HB (100073368), PDLIM1 (100069996), SYNCRIP (100074128), NISCH (100068156) and PRSS23 (100002084).

SDS–PAGE and quantitative western blotting

Protein extracts were separated under denaturing conditions by SDS–PAGE and transferred to nitrocellulose membranes. Membranes were blocked for 1 h at room temperature with blocking buffer (LI-COR Biosciences) and then incubated overnight at 4°C with the appropriate primary antibody diluted in blocking buffer. Membranes were washed with PBS and then incubated with the appropriate fluorophore-conjugated secondary antibody diluted 1:5,000 in blocking buffer for 30 min. Membranes were washed in the dark and then scanned using an Odyssey infrared imaging system (LI-COR Biosciences). Band intensity was determined by digital densitometric analysis using the Odyssey software.

Sample preparation for light microscopy

If not indicated otherwise, cells were plated on high tolerance glass-bottom dishes (MatTek Corporation, coverslip #1.7) pre-coated first with Poly-L lysine (10 μg/ml, 1 h at 37°C) and then with bovine plasma fibronectin (10 μg/ml, 2 h at 37°C).

To generate the filopodia map, U2OS cells transiently expressing a GFP-tagged protein of interest (POI) and MYO10-mScarlet were plated for 2 h on fibronectin-coated glass-bottom dishes. Samples were fixed and permeabilised simultaneously using a solution of 4% (wt/vol) PFA and 0.25% (vol/vol) Triton X-100 for 10 min. Cells were then washed with PBS, quenched using a solution of 1 M glycine for 30 min, and incubated with SiR-actin (100 nM in PBS) at 4°C until imaging (minimum length of staining, overnight at 4°C; maximum length, 1 week). Just before imaging, samples were washed three times in PBS and mounted in vectashield (Vectorlabs).

To stain endogenous proteins, U2OS cells transiently expressing MYO10-mScarlet were plated on fibronectin-coated glass-bottom dishes for 2h. Samples were fixed and permeabilised simultaneously using a solution of 4% (wt/vol) PFA and 0.25% (vol/vol) Triton X-100 for 10 min. Cells were then washed with PBS, quenched using a solution of 1 M glycine for 30 min, and incubated with the primary antibody for 1 h (1:100). After three washes, cells were incubated with a secondary antibody for 1 h (1:100). Samples were then washes three times and stored in PBS or in PBS containing an actin stain (as indicated) at 4°C until imaging. Just before imaging, samples were washed three times in PBS and mounted in vectashield (Vectorlabs).

For the filopodia formation assays, cells expressing human MYO10-GFP or MYO10-mScarlet were plated for 2 h in full medium either on glass-bottom dishes (MatTek Corporation) or on polyacrylamide gels of defined stiffness (Matrigen; 0.5 kPa, soft, SV3510-EC-0.5; 50 kPa, stiff, SV3510-EC-50) precoated with fibronectin (10 μg/ml). Cells were then fixed using PFA, washed with PBS, permeabilized and stained using phalloidin. Images were acquired using either an SDC microscope (100x objective) or an Airyscan confocal microscope (long-working-distance 63 × water objective). The number of filopodia per cell was manually scored using Fiji [80, 84]. Filopodia length were measured automatically using a custom-made Fiji plug-in as previously described [15].

Live cell imaging

All live-cell imaging experiments were performed in normal growth media, supplemented with 50 mM HEPES, at 37°C and in the presence of 5% CO2.

Dynamics of PI(3,4)P2, PXN, TLN1, ITGA5 and FMNL3 were recorded using an Airyscan microscope and a 40x objective.

To study the role of BCAR1 in filopodia stability, U2OS cells expressing MYO10-GFP were plated for at least 2 h on fibronectin before the start of live imaging (pictures taken every 5 s at 37°C, on an Airyscan microscope using a 40x objective). To study the role of extracellular stiffness in filopodia stability, U2OS cells expressing MYO10-GFP were plated for at least 2 h on polyacrylamide gels of defined stiffness (Matrigen; 0.5 kPa, soft, SV3510-EC-0.5; 50 kPa, stiff, SV3510-EC-50) precoated with fibronectin (10 μg/ml) before the start of live imaging (pictures taken every 5 s at 37°C, on an Airyscan microscope using a long-working-distance 63 × water objective). Filopodia lifetimes were then measured by identifying and tracking all MYO10 spots using the Fiji plugin TrackMate [81]. In Trackmate, the LoG detector (estimated bob diameter = 0.8 μm; threshold = 20; subpixel localization enabled) and the simple LAP tracker (linking max distance = 1 μm; gap-closing max distance = 1 μm; gap-closing max frame gap = 0) were used.

Traction force microscopy (TFM)

TFM analysis is performed by recording the position of fluorescent beads that are incorporated into a deformable gel in the presence and in the absence of cells. Bead displacement is then used to calculate the amount of force generated by cells onto the substrate.

Hydrogels used for TFM analyses were prepared as follows: glass-bottom dishes (MatTek, P35G-1.0-14-C) were treated with a solution of bind-silane (Sigma-Aldrich, Silane A-174) for 15 min at RT, washed once with 95% EtOH, twice with mQH2O before being left to dry. A pre-mixture composed of 94 μl of 40% acrylamide (Sigma, A4058), 50 μl of 2% N, N′-Methylenebisacrylamide solution (Sigma, M1533), and 356 μl of PBS was prepared (yielding a stiffness of ∼10kPa [59]). followed by addition of 3.4 μL of sonicated FluoSpheres (505/515) (Life Technologies, F881). The pre-mixture was then vortexed briefly, and 1/500 of TEMED (Sigma, T9281) and 1/100 APS 10% was added immediately before 11.7 μL of the mixture was pipetted onto the glass plates. A round 13 mm coverslip was carefully placed on top of the drop, ensuring that a thin layer of liquid remained between the two glass surfaces, and incubated for 30 min. The plate was then submerged in PBS and the glass coverslip was carefully removed. For functionalization, Sulfo-SANPAH (Thermo Scientific, 22589) 0.2 mg/ml and N-(3-Dimethylaminopropyl)-N’ ethylcarbodiimide hydrochloride (EDC) (Sigma, 03450) 2 mg/ml in 50 mM HEPES were added onto the gels and incubated for 30 min at RT with gentle agitation. Gels were placed into a UV-chamber for 10 min without cover for polymerization, washed three times with PBS and then coated with fibronectin at +4°C overnight.

Cells were plated onto TFM plates 2-4 h before the experiment. Cells were imaged live using a spinning disk confocal microscope (long-working-distance 63 × water objective). A fluorescence image of the beads (Excitation, 488 nm; Detection, 500–550 nm) and a phase-contrast image of the cells were recorded. To acquire a reference image (beads position in absence of cells), cells were removed by adding 20% SDS. Bead displacement was analyzed using a MATLAB (MathWorks) software package, kindly provided by Timo Betz (University of Münster), that employs a correlation algorithm previously described [82]. Traction stresses were determined using the same MATLAB software package following the Fourier transform traction force algorithm [85].

Microscopy setup

The structured illumination microscope (SIM) used was DeltaVision OMX v4 (GE Healthcare Life Sciences) fitted with a 60 × Plan-Apochromat objective lens, 1.42 NA (immersion oil RI of 1.516) used in SIM illumination mode (five phases × three rotations). Emitted light was collected on a front illuminated pco.edge sCMOS (pixel size 6.5 μm, readout speed 95 MHz; PCO AG) controlled by SoftWorx.

The spinning disk microscope used was a Marianas spinning disk imaging system with a Yokogawa CSU-1 scanning unit on an inverted Zeiss Axio Observer Z1 microscope controlled by SlideBook 6 (Intelligent Imaging Innovations). Objectives used were a long-working-distance 63 × water (NA 1.15 water, LDC-Apochromat, M27) objective or a 100 × (NA 1.4 oil, Plan-Apochromat, M27) objective. Images were acquired using either an Orca Flash4 sCMOS camera (chip size 2,048 × 2,048; Binning 2x2 enabled; Hamamatsu Photonics) or an Evolve 512 EMCCD camera (chip size 512 × 512; Photometrics).

The confocal microscope used was a laser scanning confocal microscope LSM880 (Zeiss) equipped with an Airyscan detector (Carl Zeiss). Objectives used were a long-working-distance 63 × water (NA 1.15 water, LDC-Apochromat, M27) or a 40x oil (1.4). The microscope was controlled using Zen Black (2.3) and the Airyscan was used in standard super-resolution mode.

Mapping of proteins within filopodia

To map the localization of each POI within filopodia, images were first processed in Fiji [80] and data analyzed using R. Briefly, in Fiji, the brightness and contrast of each image was automatically adjusted using, as an upper maximum, the brightest cellular structure labeled in the field of view. In Fiji, line intensity profiles (1 pixel width) were manually drawn from filopodium tip to base (defined by the intersection of the filopodium and the lamellipodium). To avoid any bias in the analysis, the intensity profile lines were drawn from a merged image of actin, MYO10, and POI. All visible filopodia in each images were analyzed and exported for further analysis (export was performed using the “Multi Plot” function). Data S1 contains the number of filopodia and cells analyzed for each POI. The Fiji script used to process the data is available as supplementary information (Data S2). For each POI, line intensity profiles were then compiled and analyzed in R. To homogenize filopodia length, each line intensity profile was binned into 40 bins (using the median value of pixels in each bin and the R function “tapply”). Using the line intensity profiles, the percentage of filopodia positive for each POI was quantified. A positive identification was defined as requiring at least three bins (out of 40), each with a minimum value of 10000 (bin values between 0-65535). The map of each POI was created by averaging hundreds of binned intensity profiles. The R script used to bin and average line intensity profiles is available as supplementary information (Data S2). The averaged binned intensity profiles of each POI is available in Data S2. The filopodia maps were then displayed as heatmaps in R (Data S2).

The length of each filopodia analyzed were directly extracted from the line intensity profiles and are plotted in Figure S1.

The preferential recruitment of core filopodia proteins to filopodia tips or shafts was assessed by calculating an enrichment ratio where the averaged intensity of a POI at filopodium tip (bin 1-6) was divided by the averaged intensity of a POI at filopodia shaft (bin 7-40). This enrichment ratio was calculated for each filopodium analyzed and the results are displayed as Tukey boxplots in Figure 4.

The functional annotation analysis (protein domain enrichments and search for known interactors) of the core filopodia proteins was performed using the DAVID platform [77].

Quantification and Statistical Analysis

The images used to generate the filopodia map were acquired from two independent experiments. All other experiments were replicated at least three times. No strategy was employed for randomization and/or stratification. No blinding or sample-size estimations were performed at any stage of the study. No data were excluded from the analyses.

The Tukey boxplots represent the median and the 25th and 75th percentiles (interquartile range); points are displayed as outliers (represented by dots) if 1.5 times above or below the interquartile range (represented by whiskers). Boxplots were generated using the online tool BoxPlotR (http://shiny.chemgrid.org/boxplotr/) [78]. Violin plots and dot plots were generated using the online tool PlotsOfData (https://huygens.science.uva.nl/PlotsOfData/) [79].

Statistical analyses were performed when appropriate, and p values are indicated in the figure legends. Unless otherwise indicated, the Student’s t test was used (unpaired, two tailed, and unequal variance, performed within LibreOffice Calc).

Data and Software Availability

Representative images highlighting the subcellular localization of each protein of interest (POI) to generate the filopodia map are available in Data S2. Data S2 also contains the scripts used for generating the map. Script 1 is the ImageJ macro used to measure and export the line intensity profiles from filopodia. Script 2 is the R code used to extract, compile, and average the line intensity profiles previously measured in ImageJ. Script 3 is the R code used to generate the filopodia map (using the “input table.csv” file) displayed in Figure 2. The “input table.csv” file contains the numerical values used to generate the filopodia map displayed in Figure 2.

Acknowledgments

We thank J. Siivonen and P. Laasola for technical assistance and M. Saari for help with the microscopes. We thank T. Balla, B. Baum, J. Bird, D. Bouvard, M. Davidson, R. Fässler, G. Giannone, J. Goedhart, B. Goult, R. Grosse, H. Higgs, R. Horwitz, M. Krause, E. Luna, J.F. Marshall, D. Mielenz, M. Parsons, A. Ross, D. Rösel, D. Sacks, G. Scita, G. Serini, H. Stenmark, J. Stow, E. Strehler, M. Welch, M. Wymann, and K. Yamada for providing reagents. The Cell Imaging Core (Turku Centre for Biotechnology, University of Turku, Åbo Akademi University and Biocenter Finland) and the Genome Biology Unit (Research Programs Unit, HiLIFE Helsinki Institute of Life Science, Faculty of Medicine, University of Helsinki, Biocenter Finland) are acknowledged for services, instrumentation, and expertise. This study has been supported by the Academy of Finland (G.J. and J.I.), Academy of Finland CoE for Translational Cancer Research (J.I.), ERC CoG grant 615258 (J.I.), Sigrid Juselius Foundation, and the Finnish Cancer Organization (J.I.). A.S. has been supported by the Turku Doctoral Program for Molecular Medicine. M.M. has been supported by the Turku Drug Research Doctoral Programme.

Author Contributions

Conceptualization, G.J.; Methodology, G.J.; Formal Analysis, G.J.; Investigation, G.J. and A.S.; Resources, G.J., R.S., E.K., and M.M.; Writing – Original Draft, G.J.; Writing – Review and Editing, G.J., J.I., and H.H.; Visualization, G.J.; Supervision, G.J. and J.I.; Funding Acquisition, G.J. and J.I.

Declaration of Interests

The authors declare no competing interests.

Published: January 10, 2019

Footnotes

Supplemental Information includes six figures, six videos, and two data files and can be found with this article online at https://doi.org/10.1016/j.cub.2018.11.053

Contributor Information

Guillaume Jacquemet, Email: guillaume.jacquemet@utu.fi.

Johanna Ivaska, Email: johanna.ivaska@utu.fi.

Supplemental Information

Document S1. Figures S1–S6
mmc1.pdf (20MB, pdf)
Data S1. Constructs Imaged to Generate the Filopodia Map, Related to Figures 1, 2, 3, 4, and 6

Data S1 highlights the various proteins / constructs imaged in this study as well as the number of filopodia analyzed to generate Figures 1, 2, 3, 4, and 7.

mmc8.xlsx (13.8KB, xlsx)
Data S2. Resources Used to Generate the Filopodia Map, Related to Figures 1, 2, 3, 4, and 6

Data S2 contains a PDF file where representative images highlight the subcellular localisation of each protein of interest (POI) to generate the filopodia map (Figure 2). Data S2 also contains the scripts used for generating the map. Script 1 is the ImageJ macro used to measure and export the line intensity profiles from filopodia. Script 2 is the R code used to extract, compile and average the line intensity profiles previously measured in ImageJ. Script 3 is the R code used to generate the filopodia map (using the “input table.csv” file) displayed in Figure 2. The “input table.csv” file contains the numerical values used to generate the filopodia map displayed in Figure 2.

mmc9.zip (43.1MB, zip)
Document S2. Article plus Supplemental Information
mmc10.pdf (24.7MB, pdf)

References

  • 1.Legate K.R., Wickström S.A., Fässler R. Genetic and cell biological analysis of integrin outside-in signaling. Genes Dev. 2009;23:397–418. doi: 10.1101/gad.1758709. [DOI] [PubMed] [Google Scholar]
  • 2.Askari J.A., Buckley P.A., Mould A.P., Humphries M.J. Linking integrin conformation to function. J. Cell Sci. 2009;122:165–170. doi: 10.1242/jcs.018556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zaidel-Bar R., Geiger B. The switchable integrin adhesome. J. Cell Sci. 2010;123:1385–1388. doi: 10.1242/jcs.066183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Horton E.R., Byron A., Askari J.A., Ng D.H.J., Millon-Frémillon A., Robertson J., Koper E.J., Paul N.R., Warwood S., Knight D. Definition of a consensus integrin adhesome and its dynamics during adhesion complex assembly and disassembly. Nat. Cell Biol. 2015;17:1577–1587. doi: 10.1038/ncb3257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Morgan M.R., Byron A., Humphries M.J., Bass M.D. Giving off mixed signals--distinct functions of alpha5beta1 and alphavbeta3 integrins in regulating cell behaviour. IUBMB Life. 2009;61:731–738. doi: 10.1002/iub.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Jansen K.A., Atherton P., Ballestrem C. Mechanotransduction at the cell-matrix interface. Semin. Cell Dev. Biol. 2017;71:75–83. doi: 10.1016/j.semcdb.2017.07.027. [DOI] [PubMed] [Google Scholar]
  • 7.Jacquemet G., Hamidi H., Ivaska J. Filopodia in cell adhesion, 3D migration and cancer cell invasion. Curr. Opin. Cell Biol. 2015;36:23–31. doi: 10.1016/j.ceb.2015.06.007. [DOI] [PubMed] [Google Scholar]
  • 8.Jacquemet G., Paatero I., Carisey A.F., Padzik A., Orange J.S., Hamidi H., Ivaska J. FiloQuant reveals increased filopodia density during breast cancer progression. J. Cell Biol. 2017;216:3387–3403. doi: 10.1083/jcb.201704045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Johnson H.E., King S.J., Asokan S.B., Rotty J.D., Bear J.E., Haugh J.M. F-actin bundles direct the initiation and orientation of lamellipodia through adhesion-based signaling. J. Cell Biol. 2015;208:443–455. doi: 10.1083/jcb.201406102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Albuschies J., Vogel V. The role of filopodia in the recognition of nanotopographies. Sci. Rep. 2013;3:1658. doi: 10.1038/srep01658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wong S., Guo W.-H., Wang Y.-L. Fibroblasts probe substrate rigidity with filopodia extensions before occupying an area. Proc. Natl. Acad. Sci. USA. 2014;111:17176–17181. doi: 10.1073/pnas.1412285111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chan C.E., Odde D.J. Traction dynamics of filopodia on compliant substrates. Science. 2008;322:1687–1691. doi: 10.1126/science.1163595. [DOI] [PubMed] [Google Scholar]
  • 13.Kim M.-C., Silberberg Y.R., Abeyaratne R., Kamm R.D., Asada H.H. Computational modeling of three-dimensional ECM-rigidity sensing to guide directed cell migration. Proc. Natl. Acad. Sci. USA. 2018;115:E390–E399. doi: 10.1073/pnas.1717230115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Zhang H., Berg J.S., Li Z., Wang Y., Lång P., Sousa A.D., Bhaskar A., Cheney R.E., Strömblad S. Myosin-X provides a motor-based link between integrins and the cytoskeleton. Nat. Cell Biol. 2004;6:523–531. doi: 10.1038/ncb1136. [DOI] [PubMed] [Google Scholar]
  • 15.Jacquemet G., Baghirov H., Georgiadou M., Sihto H., Peuhu E., Cettour-Janet P., He T., Perälä M., Kronqvist P., Joensuu H., Ivaska J. L-type calcium channels regulate filopodia stability and cancer cell invasion downstream of integrin signalling. Nat. Commun. 2016;7:13297. doi: 10.1038/ncomms13297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kanchanawong P., Shtengel G., Pasapera A.M., Ramko E.B., Davidson M.W., Hess H.F., Waterman C.M. Nanoscale architecture of integrin-based cell adhesions. Nature. 2010;468:580–584. doi: 10.1038/nature09621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lagarrigue F., Vikas Anekal P., Lee H.-S., Bachir A.I., Ablack J.N., Horwitz A.F., Ginsberg M.H. A RIAM/lamellipodin-talin-integrin complex forms the tip of sticky fingers that guide cell migration. Nat. Commun. 2015;6:8492. doi: 10.1038/ncomms9492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Harris E.S., Gauvin T.J., Heimsath E.G., Higgs H.N. Assembly of filopodia by the formin FRL2 (FMNL3) Cytoskeleton (Hoboken) 2010;67:755–772. doi: 10.1002/cm.20485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Krause M., Leslie J.D., Stewart M., Lafuente E.M., Valderrama F., Jagannathan R., Strasser G.A., Rubinson D.A., Liu H., Way M. Lamellipodin, an Ena/VASP ligand, is implicated in the regulation of lamellipodial dynamics. Dev. Cell. 2004;7:571–583. doi: 10.1016/j.devcel.2004.07.024. [DOI] [PubMed] [Google Scholar]
  • 20.Rottner K., Behrendt B., Small J.V., Wehland J. VASP dynamics during lamellipodia protrusion. Nat. Cell Biol. 1999;1:321–322. doi: 10.1038/13040. [DOI] [PubMed] [Google Scholar]
  • 21.Yang C., Czech L., Gerboth S., Kojima S., Scita G., Svitkina T. Novel roles of formin mDia2 in lamellipodia and filopodia formation in motile cells. PLoS Biol. 2007;5:e317. doi: 10.1371/journal.pbio.0050317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Vignjevic D., Kojima S., Aratyn Y., Danciu O., Svitkina T., Borisy G.G. Role of fascin in filopodial protrusion. J. Cell Biol. 2006;174:863–875. doi: 10.1083/jcb.200603013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Maekawa M., Fairn G.D. Molecular probes to visualize the location, organization and dynamics of lipids. J. Cell Sci. 2014;127:4801–4812. doi: 10.1242/jcs.150524. [DOI] [PubMed] [Google Scholar]
  • 24.Liu P.-S., Jong T.-H., Maa M.-C., Leu T.-H. The interplay between Eps8 and IRSp53 contributes to Src-mediated transformation. Oncogene. 2010;29:3977–3989. doi: 10.1038/onc.2010.144. [DOI] [PubMed] [Google Scholar]
  • 25.Gardel M.L., Schneider I.C., Aratyn-Schaus Y., Waterman C.M. Mechanical integration of actin and adhesion dynamics in cell migration. Annu. Rev. Cell Dev. Biol. 2010;26:315–333. doi: 10.1146/annurev.cellbio.011209.122036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zaidel-Bar R., Ballestrem C., Kam Z., Geiger B. Early molecular events in the assembly of matrix adhesions at the leading edge of migrating cells. J. Cell Sci. 2003;116:4605–4613. doi: 10.1242/jcs.00792. [DOI] [PubMed] [Google Scholar]
  • 27.Kuo J.-C., Han X., Hsiao C.-T., Yates J.R., 3rd, Waterman C.M. Analysis of the myosin-II-responsive focal adhesion proteome reveals a role for β-Pix in negative regulation of focal adhesion maturation. Nat. Cell Biol. 2011;13:383–393. doi: 10.1038/ncb2216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Azatov M., Goicoechea S.M., Otey C.A., Upadhyaya A. The actin crosslinking protein palladin modulates force generation and mechanosensitivity of tumor associated fibroblasts. Sci. Rep. 2016;6:28805. doi: 10.1038/srep28805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Pasapera A.M., Schneider I.C., Rericha E., Schlaepfer D.D., Waterman C.M. Myosin II activity regulates vinculin recruitment to focal adhesions through FAK-mediated paxillin phosphorylation. J. Cell Biol. 2010;188:877–890. doi: 10.1083/jcb.200906012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lawson C., Lim S.-T.S.-T., Uryu S., Chen X.L., Calderwood D.A., Schlaepfer D.D. FAK promotes recruitment of talin to nascent adhesions to control cell motility. J. Cell Biol. 2012;196:223–232. doi: 10.1083/jcb.201108078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Swaminathan V., Fischer R.S., Waterman C.M. The FAK-Arp2/3 interaction promotes leading edge advance and haptosensing by coupling nascent adhesions to lamellipodia actin. Mol. Biol. Cell. 2016;27:1085–1100. doi: 10.1091/mbc.E15-08-0590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.He K., Sakai T., Tsukasaki Y., Watanabe T.M., Ikebe M. Myosin X is recruited to nascent focal adhesions at the leading edge and induces multi-cycle filopodial elongation. Sci. Rep. 2017;7:13685. doi: 10.1038/s41598-017-06147-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sawada Y., Tamada M., Dubin-Thaler B.J., Cherniavskaya O., Sakai R., Tanaka S., Sheetz M.P. Force sensing by mechanical extension of the Src family kinase substrate p130Cas. Cell. 2006;127:1015–1026. doi: 10.1016/j.cell.2006.09.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Donato D.M., Ryzhova L.M., Meenderink L.M., Kaverina I., Hanks S.K. Dynamics and mechanism of p130Cas localization to focal adhesions. J. Biol. Chem. 2010;285:20769–20779. doi: 10.1074/jbc.M109.091207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang Y., McNiven M.A. Invasive matrix degradation at focal adhesions occurs via protease recruitment by a FAK-p130Cas complex. J. Cell Biol. 2012;196:375–385. doi: 10.1083/jcb.201105153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Janoštiak R., Pataki A.C., Brábek J., Rösel D. Mechanosensors in integrin signaling: the emerging role of p130Cas. Eur. J. Cell Biol. 2014;93:445–454. doi: 10.1016/j.ejcb.2014.07.002. [DOI] [PubMed] [Google Scholar]
  • 37.Braniš J., Pataki C., Spörrer M., Gerum R.C., Mainka A., Cermak V., Goldmann W.H., Fabry B., Brabek J., Rosel D. The role of focal adhesion anchoring domains of CAS in mechanotransduction. Sci. Rep. 2017;7:46233. doi: 10.1038/srep46233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Humphries J.D., Byron A., Bass M.D., Craig S.E., Pinney J.W., Knight D., Humphries M.J. Proteomic analysis of integrin-associated complexes identifies RCC2 as a dual regulator of Rac1 and Arf6. Sci. Signal. 2009;2:ra51. doi: 10.1126/scisignal.2000396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Miki H., Sasaki T., Takai Y., Takenawa T. Induction of filopodium formation by a WASP-related actin-depolymerizing protein N-WASP. Nature. 1998;391:93–96. doi: 10.1038/34208. [DOI] [PubMed] [Google Scholar]
  • 40.Lee K., Gallop J.L., Rambani K., Kirschner M.W. Self-assembly of filopodia-like structures on supported lipid bilayers. Science. 2010;329:1341–1345. doi: 10.1126/science.1191710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Plantard L., Arjonen A., Lock J.G., Nurani G., Ivaska J., Strömblad S. PtdIns(3,4,5)P3 is a regulator of myosin-X localization and filopodia formation. J. Cell Sci. 2010;123:3525–3534. doi: 10.1242/jcs.069609. [DOI] [PubMed] [Google Scholar]
  • 42.Sharma V.P., Eddy R., Entenberg D., Kai M., Gertler F.B., Condeelis J. Tks5 and SHIP2 regulate invadopodium maturation, but not initiation, in breast carcinoma cells. Curr. Biol. 2013;23:2079–2089. doi: 10.1016/j.cub.2013.08.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rajadurai C.V., Havrylov S., Coelho P.P., Ratcliffe C.D.H., Zaoui K., Huang B.H., Monast A., Chughtai N., Sangwan V., Gertler F.B. 5′-inositol phosphatase SHIP2 recruits Mena to stabilize invadopodia for cancer cell invasion. J. Cell Biol. 2016;214:719–734. doi: 10.1083/jcb.201501003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Daste F., Walrant A., Holst M.R., Gadsby J.R., Mason J., Lee J.-E., Brook D., Mettlen M., Larsson E., Lee S.F. Control of actin polymerization via the coincidence of phosphoinositides and high membrane curvature. J. Cell Biol. 2017;216:3745–3765. doi: 10.1083/jcb.201704061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Morse E.M., Brahme N.N., Calderwood D.A. Integrin cytoplasmic tail interactions. Biochemistry. 2014;53:810–820. doi: 10.1021/bi401596q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hu W., Wehrle-Haller B., Vogel V. Maturation of filopodia shaft adhesions is upregulated by local cycles of lamellipodia advancements and retractions. PLoS ONE. 2014;9:e107097. doi: 10.1371/journal.pone.0107097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jacquemet G., Green D.M., Bridgewater R.E., von Kriegsheim A., Humphries M.J., Norman J.C., Caswell P.T. RCP-driven α5β1 recycling suppresses Rac and promotes RhoA activity via the RacGAP1-IQGAP1 complex. J. Cell Biol. 2013;202:917–935. doi: 10.1083/jcb.201302041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Paul N.R., Allen J.L., Chapman A., Morlan-Mairal M., Zindy E., Jacquemet G., Fernandez del Ama L., Ferizovic N., Green D.M., Howe J.D. α5β1 integrin recycling promotes Arp2/3-independent cancer cell invasion via the formin FHOD3. J. Cell Biol. 2015;210:1013–1031. doi: 10.1083/jcb.201502040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Bornschlögl T. How filopodia pull: what we know about the mechanics and dynamics of filopodia. Cytoskeleton (Hoboken) 2013;70:590–603. doi: 10.1002/cm.21130. [DOI] [PubMed] [Google Scholar]
  • 50.Bornschlögl T., Romero S., Vestergaard C.L., Joanny J.-F., Van Nhieu G.T., Bassereau P. Filopodial retraction force is generated by cortical actin dynamics and controlled by reversible tethering at the tip. Proc. Natl. Acad. Sci. USA. 2013;110:18928–18933. doi: 10.1073/pnas.1316572110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Leijnse N., Oddershede L.B., Bendix P.M. Helical buckling of actin inside filopodia generates traction. Proc. Natl. Acad. Sci. USA. 2015;112:136–141. doi: 10.1073/pnas.1411761112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Alieva N.O., Efremov A.K., Hu S., Oh D., Chen Z., Natarajan M., Ong H.T., Jegou A., Romet-Lemonne G., Groves J.T. Force dependence of filopodia adhesion: involvement of myosin II and formins. bioRxiv. 2018 doi: 10.1038/s41467-019-10964-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Yao M., Goult B.T., Klapholz B., Hu X., Toseland C.P., Guo Y., Cong P., Sheetz M.P., Yan J. The mechanical response of talin. Nat. Commun. 2016;7:11966. doi: 10.1038/ncomms11966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hotta K., Ranganathan S., Liu R., Wu F., Machiyama H., Gao R., Hirata H., Soni N., Ohe T., Hogue C.W.V. Biophysical properties of intrinsically disordered p130Cas substrate domain--implication in mechanosensing. PLoS Comput. Biol. 2014;10:e1003532. doi: 10.1371/journal.pcbi.1003532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kirsch K.H., Georgescu M.M., Hanafusa H. Direct binding of p130(Cas) to the guanine nucleotide exchange factor C3G. J. Biol. Chem. 1998;273:25673–25679. doi: 10.1074/jbc.273.40.25673. [DOI] [PubMed] [Google Scholar]
  • 56.Gotoh T., Cai D., Tian X., Feig L.A., Lerner A. p130Cas regulates the activity of AND-34, a novel Ral, Rap1, and R-Ras guanine nucleotide exchange factor. J. Biol. Chem. 2000;275:30118–30123. doi: 10.1074/jbc.M003074200. [DOI] [PubMed] [Google Scholar]
  • 57.Miller F.R., Santner S.J., Tait L., Dawson P.J. MCF10DCIS.com xenograft model of human comedo ductal carcinoma in situ. J. Natl. Cancer Inst. 2000;92:1185–1186. doi: 10.1093/jnci/92.14.1185a. [DOI] [PubMed] [Google Scholar]
  • 58.Muharram G., Sahgal P., Korpela T., De Franceschi N., Kaukonen R., Clark K., Tulasne D., Carpén O., Ivaska J. Tensin-4-dependent MET stabilization is essential for survival and proliferation in carcinoma cells. Dev. Cell. 2014;29:421–436. doi: 10.1016/j.devcel.2014.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Georgiadou M., Lilja J., Jacquemet G., Guzmán C., Rafaeva M., Alibert C., Yan Y., Sahgal P., Lerche M., Manneville J.-B. AMPK negatively regulates tensin-dependent integrin activity. J. Cell Biol. 2017;216:1107–1121. doi: 10.1083/jcb.201609066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rantala J.K., Pouwels J., Pellinen T., Veltel S., Laasola P., Mattila E., Potter C.S., Duffy T., Sundberg J.P., Kallioniemi O. SHARPIN is an endogenous inhibitor of β1-integrin activation. Nat. Cell Biol. 2011;13:1315–1324. doi: 10.1038/ncb2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nevo J., Mattila E., Pellinen T., Yamamoto D.L., Sara H., Iljin K., Kallioniemi O., Bono P., Heikkilä P., Joensuu H. Mammary-derived growth inhibitor alters traffic of EGFR and induces a novel form of cetuximab resistance. Clin. Cancer Res. 2009;15:6570–6581. doi: 10.1158/1078-0432.CCR-09-0773. [DOI] [PubMed] [Google Scholar]
  • 62.Avramidou A., Kroczek C., Lang C., Schuh W., Jäck H.-M., Mielenz D. The novel adaptor protein Swiprosin-1 enhances BCR signals and contributes to BCR-induced apoptosis. Cell Death Differ. 2007;14:1936–1947. doi: 10.1038/sj.cdd.4402206. [DOI] [PubMed] [Google Scholar]
  • 63.Grikscheit K., Frank T., Wang Y., Grosse R. Junctional actin assembly is mediated by Formin-like 2 downstream of Rac1. J. Cell Biol. 2015;209:367–376. doi: 10.1083/jcb.201412015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Belyantseva I.A., Boger E.T., Naz S., Frolenkov G.I., Sellers J.R., Ahmed Z.M., Griffith A.J., Friedman T.B. Myosin-XVa is required for tip localization of whirlin and differential elongation of hair-cell stereocilia. Nat. Cell Biol. 2005;7:148–156. doi: 10.1038/ncb1219. [DOI] [PubMed] [Google Scholar]
  • 65.Sun Z., Tseng H.-Y., Tan S., Senger F., Kurzawa L., Dedden D., Mizuno N., Wasik A.A., Thery M., Dunn A.R., Fässler R. Kank2 activates talin, reduces force transduction across integrins and induces central adhesion formation. Nat. Cell Biol. 2016;18:941–953. doi: 10.1038/ncb3402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gillooly D.J., Morrow I.C., Lindsay M., Gould R., Bryant N.J., Gaullier J.M., Parton R.G., Stenmark H. Localization of phosphatidylinositol 3-phosphate in yeast and mammalian cells. EMBO J. 2000;19:4577–4588. doi: 10.1093/emboj/19.17.4577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chertkova A.O., Mastop M., Postma M., van Bommel N., van der Niet S., Batenburg K.L., Joosen L., Gadella T.W.J., Okada Y., Goedhart J. Robust and bright genetically encoded fluorescent markers for highlighting structures and compartments in mammalian cells. bioRxiv. 2017 [Google Scholar]
  • 68.Ren J.-G., Li Z., Crimmins D.L., Sacks D.B. Self-association of IQGAP1: characterization and functional sequelae. J. Biol. Chem. 2005;280:34548–34557. doi: 10.1074/jbc.M507321200. [DOI] [PubMed] [Google Scholar]
  • 69.Burnette D.T., Shao L., Ott C., Pasapera A.M., Fischer R.S., Baird M.A., Der Loughian C., Delanoe-Ayari H., Paszek M.J., Davidson M.W. A contractile and counterbalancing adhesion system controls the 3D shape of crawling cells. J. Cell Biol. 2014;205:83–96. doi: 10.1083/jcb.201311104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Liu F., Wagner S., Campbell R.B., Nickerson J.A., Schiffer C.A., Ross A.H. PTEN enters the nucleus by diffusion. J. Cell. Biochem. 2005;96:221–234. doi: 10.1002/jcb.20525. [DOI] [PubMed] [Google Scholar]
  • 71.Paszek M.J., DuFort C.C., Rubashkin M.G., Davidson M.W., Thorn K.S., Liphardt J.T., Weaver V.M. Scanning angle interference microscopy reveals cell dynamics at the nanoscale. Nat. Methods. 2012;9:825–827. doi: 10.1038/nmeth.2077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Miranda K.C., Khromykh T., Christy P., Le T.L., Gottardi C.J., Yap A.S., Stow J.L., Teasdale R.D. A dileucine motif targets E-cadherin to the basolateral cell surface in Madin-Darby canine kidney and LLC-PK1 epithelial cells. J. Biol. Chem. 2001;276:22565–22572. doi: 10.1074/jbc.M101907200. [DOI] [PubMed] [Google Scholar]
  • 73.Hertzog M., Milanesi F., Hazelwood L., Disanza A., Liu H., Perlade E., Malabarba M.G., Pasqualato S., Maiolica A., Confalonieri S. Molecular basis for the dual function of Eps8 on actin dynamics: bundling and capping. PLoS Biol. 2010;8:e1000387. doi: 10.1371/journal.pbio.1000387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hammond G.R.V., Machner M.P., Balla T. A novel probe for phosphatidylinositol 4-phosphate reveals multiple pools beyond the Golgi. J. Cell Biol. 2014;205:113–126. doi: 10.1083/jcb.201312072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bennett R.D., Strehler E.E. Calmodulin-like protein enhances myosin-10 translation. Biochem. Biophys. Res. Commun. 2008;369:654–659. doi: 10.1016/j.bbrc.2008.02.056. [DOI] [PubMed] [Google Scholar]
  • 76.Welch M.D., DePace A.H., Verma S., Iwamatsu A., Mitchison T.J. The human Arp2/3 complex is composed of evolutionarily conserved subunits and is localized to cellular regions of dynamic actin filament assembly. J. Cell Biol. 1997;138:375–384. doi: 10.1083/jcb.138.2.375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Huang da W., Sherman B.T., Lempicki R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2009;4:44–57. doi: 10.1038/nprot.2008.211. [DOI] [PubMed] [Google Scholar]
  • 78.Spitzer M., Wildenhain J., Rappsilber J., Tyers M. BoxPlotR: a web tool for generation of box plots. Nat. Methods. 2014;11:121–122. doi: 10.1038/nmeth.2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Postma M., Goedhart J. PlotsOfData - a web app for visualizing data together with its summaries. bioRxiv. 2018 doi: 10.1371/journal.pbio.3000202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Schindelin J., Arganda-Carreras I., Frise E., Kaynig V., Longair M., Pietzsch T., Preibisch S., Rueden C., Saalfeld S., Schmid B. Fiji: an open-source platform for biological-image analysis. Nat. Methods. 2012;9:676–682. doi: 10.1038/nmeth.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tinevez J.-Y., Perry N., Schindelin J., Hoopes G.M., Reynolds G.D., Laplantine E., Bednarek S.Y., Shorte S.L., Eliceiri K.W. TrackMate: an open and extensible platform for single-particle tracking. Methods. 2017;115:80–90. doi: 10.1016/j.ymeth.2016.09.016. [DOI] [PubMed] [Google Scholar]
  • 82.Betz T., Koch D., Lu Y.-B., Franze K., Käs J.A. Growth cones as soft and weak force generators. Proc. Natl. Acad. Sci. USA. 2011;108:13420–13425. doi: 10.1073/pnas.1106145108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bindels D.S., Haarbosch L., van Weeren L., Postma M., Wiese K.E., Mastop M., Aumonier S., Gotthard G., Royant A., Hink M.A., Gadella T.W., Jr. mScarlet: a bright monomeric red fluorescent protein for cellular imaging. Nat. Methods. 2017;14:53–56. doi: 10.1038/nmeth.4074. [DOI] [PubMed] [Google Scholar]
  • 84.Rueden C.T., Schindelin J., Hiner M.C., DeZonia B.E., Walter A.E., Arena E.T., Eliceiri K.W. ImageJ2: ImageJ for the next generation of scientific image data. BMC Bioinformatics. 2017;18:529. doi: 10.1186/s12859-017-1934-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Butler J.P., Tolić-Nørrelykke I.M., Fabry B., Fredberg J.J. Traction fields, moments, and strain energy that cells exert on their surroundings. Am. J. Physiol. Cell Physiol. 2002;282:C595–C605. doi: 10.1152/ajpcell.00270.2001. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Video S1. PI(3,4)P2 Dynamics in Filopodia, Related to Figure 3

U2OS cells expressing MYO10-mScarlet, GFP-TAPP-PH and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope. The yellow square highlights a ROI, which is magnified.

Download video file (4.7MB, mp4)
Video S2. Paxillin Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet, PXN-GFP and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope. The white square highlights a ROI, which is magnified.

Download video file (14.7MB, mp4)
Video S3. BCAR1 Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet, BCAR1-GFP and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope.

Download video file (4.3MB, mp4)
Video S4. TLN1 Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet, TLN1-GFP and lifeact-mTurquoise2 were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope.

Download video file (8.3MB, mp4)
Video S5. ITGA5 Dynamics in Filopodia, Related to Figure 5

U2OS cells expressing MYO10-mScarlet and ITGA5-GFP were plated on fibronectin for 2 h before being imaged live using an Airyscan confocal microscope.

Download video file (6.5MB, mp4)
Video S6. Dynamics of MYO10- and FMNL3-Induced Filopodia, Related to Figure 7

U2OS cells expressing lifeact-mTurquoise2 and MYO10-GFP or lifeact-mTurquoise2 and FMNL3-GFP were plated on fibronectin and imaged live using an Airyscan confocal microscope.

Download video file (7MB, mp4)
Document S1. Figures S1–S6
mmc1.pdf (20MB, pdf)
Data S1. Constructs Imaged to Generate the Filopodia Map, Related to Figures 1, 2, 3, 4, and 6

Data S1 highlights the various proteins / constructs imaged in this study as well as the number of filopodia analyzed to generate Figures 1, 2, 3, 4, and 7.

mmc8.xlsx (13.8KB, xlsx)
Data S2. Resources Used to Generate the Filopodia Map, Related to Figures 1, 2, 3, 4, and 6

Data S2 contains a PDF file where representative images highlight the subcellular localisation of each protein of interest (POI) to generate the filopodia map (Figure 2). Data S2 also contains the scripts used for generating the map. Script 1 is the ImageJ macro used to measure and export the line intensity profiles from filopodia. Script 2 is the R code used to extract, compile and average the line intensity profiles previously measured in ImageJ. Script 3 is the R code used to generate the filopodia map (using the “input table.csv” file) displayed in Figure 2. The “input table.csv” file contains the numerical values used to generate the filopodia map displayed in Figure 2.

mmc9.zip (43.1MB, zip)
Document S2. Article plus Supplemental Information
mmc10.pdf (24.7MB, pdf)

Data Availability Statement

Representative images highlighting the subcellular localization of each protein of interest (POI) to generate the filopodia map are available in Data S2. Data S2 also contains the scripts used for generating the map. Script 1 is the ImageJ macro used to measure and export the line intensity profiles from filopodia. Script 2 is the R code used to extract, compile, and average the line intensity profiles previously measured in ImageJ. Script 3 is the R code used to generate the filopodia map (using the “input table.csv” file) displayed in Figure 2. The “input table.csv” file contains the numerical values used to generate the filopodia map displayed in Figure 2.

RESOURCES