Secreted exosomes induce filopodia formation

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife Assessment

    The authors identify a novel relationship between exosome secretion and filopodia formation that has implications for cancer cell metastasis and neuronal synapse formation. Further, they identify the exosomal cargo, THSD7A, as a regulator of this process. The data presented is convincing, and represents an important advancement in our understanding of how these two biological processes are linked and play roles in regulating cell migration and cell-cell communication.

This article has been Reviewed by the following groups

Read the full article

Abstract

Filopodia are dynamic adhesive cytoskeletal structures that are critical for directional sensing, polarization, cell-cell adhesion, and migration of diverse cell types. Filopodia are also critical for neuronal synapse formation. While dynamic rearrangement of the actin cytoskeleton is known to be critical for filopodia biogenesis, little is known about the upstream extracellular signals. Here, we identify secreted exosomes as potent regulators of filopodia formation. Inhibition of exosome secretion inhibited the formation and stabilization of filopodia in both cancer cells and neurons and inhibited subsequent synapse formation by neurons. Rescue experiments with purified small and large extracellular vesicles (EVs) identified exosome-enriched small EVs (SEVs) as having potent filopodia-inducing activity. Proteomic analyses of cancer cell-derived SEVs identified the TGF-β family coreceptor endoglin as a key SEV-enriched cargo that regulates filopodia. Cancer cell endoglin levels also affected filopodia-dependent behaviors, including metastasis of cancer cells in chick embryos and 3D migration in collagen gels. As neurons do not express endoglin, we performed a second proteomics experiment to identify SEV cargoes regulated by endoglin that might promote filopodia in both cell types. We discovered a single SEV cargo that was altered in endoglin-KD cancer SEVs, the transmembrane protein Thrombospondin Type 1 Domain Containing 7A (THSD7A). We further found that both cancer cell and neuronal SEVs carry THSD7A and that add-back of purified THSD7A is sufficient to rescue filopodia defects of both endoglin-KD cancer cells and exosome-inhibited neurons. We also find that THSD7A induces filopodia formation through activation of the Rho GTPase, Cdc42. These findings suggest a new model for filopodia formation, triggered by exosomes carrying THSD7A.

Article activity feed

  1. eLife Assessment

    The authors identify a novel relationship between exosome secretion and filopodia formation that has implications for cancer cell metastasis and neuronal synapse formation. Further, they identify the exosomal cargo, THSD7A, as a regulator of this process. The data presented is convincing, and represents an important advancement in our understanding of how these two biological processes are linked and play roles in regulating cell migration and cell-cell communication.

  2. Reviewer #1 (Public review):

    Summary:

    The study significantly advances our understanding of how exosomes regulate filopodia formation. Filopodia play crucial roles in cell movement, polarization, directional sensing, and neuronal synapse formation. McAtee et al. demonstrated that exosomes, particularly those enriched with the protein THSD7A, play a pivotal role in promoting filopodia formation through Cdc42 in cancer cells and neurons. This discovery unveils a new extracellular mechanism through which cells can control their cytoskeletal dynamics and interaction with their surroundings. The study employs a combination of rescue experiments, live-cell imaging, cell culture, and proteomic analyses to thoroughly investigate the role of exosomes and THSD7A in filopodia formation in cancer cells and neurons. These findings offer valuable insights into fundamental biological processes of cell movement and communication and have potential implications for understanding cancer metastasis and neuronal development.

    Weaknesses:

    The conclusions of this study are in most cases supported by data, but some aspects of data analysis need to be better clarified and elaborated. Some conclusions need to be better stated and according to the data observed.

  3. Reviewer #2 (Public review):

    Summary:

    The authors show that small EVs trigger the formation of filopodia in both cancer cells and neurons. They go on to show that two cargo proteins, endoglin, and THSD7A, are important for this process. This possibly occurs by activating the Rho-family GTPase CDC42.

    Strengths:

    The EV work is quite strong and convincing. The proteomics work is well executed and carefully analyzed. I was particularly impressed with the chick metastasis assay that added strong evidence of in vivo relevance.

    Weaknesses:

    The weakest part of the paper is the Cdc42 work at the end of the paper. It is incomplete and not terribly convincing. This part of the paper needs to be improved significantly

  4. Reviewer #3 (Public review):

    Summary:

    The authors identify a novel relationship between exosome secretion and filopodia formation in cancer cells and neurons. They observe that multivesicular endosomes (MVE)-plasma membrane (PM) fusion is associated with filopodia formation in HT1080 cells and that MVEs are present in filopodia in primary neurons. Using overexpression and knockdown (KD) of Rab27/HRS in HT1080 cells, melanoma cells, and/or primary rat neurons, they found that decreasing exosome secretion reduces filopodia formation, while Rab27 overexpression leads to the opposite result. Furthermore, the decreased filopodia formation is rescued in the Rab27a/HRS KD melanoma cells by the addition of small extracellular vesicles (EVs) but not large EVs purified from control cells. The authors identify endoglin as a protein unique to small EVs secreted by cancer cells when compared to large EVs. KD of endoglin reduces filopodia formation and this is rescued by the addition of small EVs from control cells and not by small EVs from endoglin KD cells. Based on the role of filopodia in cancer metastasis, the authors then investigate the role of endoglin in cancer cell metastasis using a chick embryo model. They find that injection of endoglin KD HT1080 cells into chick embryos gives rise to less metastasis compared to control cells - a phenotype that is rescued by the co-injection of small EVs from control cells. Using quantitative mass spectrometry analysis, they find that thrombospondin type 1 domain containing 7a protein (THSD7A) is downregulated in small EVs from endoglin KD melanoma cells compared to those from control cells. They also report that THSD7A is more abundant in endoglin KD cell lysate compared to control HT1080 cells and less abundant in small EVs from endoglin KD cells compared to control cells, indicating a trafficking defect. Indeed, using immunofluorescence microscopy, the authors observe THSD7A-mScarlet accumulation in CD63-positive structures in endoglin KD HT1080 cells, compared to control cells. Finally, the authors determine that exosome-secreted THSD7A induces filopodia formation in a Cdc42-dependent mechanism.

    Strengths:

    (1) While exosomes are known to play a role in cell migration and autocrine signaling, the relationship between exosome secretion and the formation of filopodia is novel.

    (2) The authors identify an exosomal cargo protein, THSD7A, which is essential for regulating this function.

    (3) The data presented provide strong evidence of a role for endoglin in the trafficking of THSD7A in exosomes.

    (4) The authors associate this process with functional significance in cancer cell metastasis and neurological synapse formation, both of which involve the formation of filopodia.

    (5) The data are presented clearly, and their interpretation appropriately explains the context and significance of the findings.

    Weaknesses:

    (1) A better characterization of the nature of the small EV population is missing:

    It is unclear why the authors chose to proceed to quantitative mass spectrometry with the bands in the Coomassie from size-separated EV samples, as there are other bands present in the small EV lane but not the large EV lane. This is important to clarify because it underlies how they were able to identify THSD7A as a unique regulator of exosome-mediated filopodia formation. Is there a reason why the total sample fractions were not compared? This would provide valuable information on the nature of the small and large EV populations.

    (2) Data analysis and quantification should be performed with increased rigor:

    a) Figure 1C - The optical and temporal resolution are insufficient to conclusively characterize the association between exosome secretion and filopodia. Specifically, the 10-second interval used in the image acquisitions is too close to the reported 20-second median time between exosome secretion and filopodia formation. Two-5 sec intervals should be used to validate this. It would also be important to correlate the percentage of filopodia events that co-occur with exosome secretion. Is this a phenomenon that occurs with most or only a small number of filopodia? Additionally, resolution with typical confocal microscopy is subpar for these analyses. TIRF microscopy would offer increased resolution to parse out secretion events. As the TIRF objective is listed in the Methods section, figure legends should mention which images were acquired using TIRF microscopy.

    b) Figure 2 - It would be important to perform further analysis to concretely determine the relationship between exosome secretion and filopodia stability. Are secretion events correlated with the stability of filopodia? Is there a positive feedback loop that causes further filopodia stability and length with increased secretion? Furthermore, is there an association between the proximity of secretion with stability? Quantification of filopodia more objectively (# of filopodia/cell) would be helpful.

    c) Figure 6 - Why use different gel conditions to detect THSD7A in small EVs from B16F1 cells vs HT1080 and neurons? Why are there two bands for THSD7A in panels C and E? It is difficult to appreciate the KD efficiency in E. The absence of a signal for THSD7A in the HT1080 shEng small EVs that show a signal for endoglin is surprising. The authors should provide rigorous quantification of the westerns from several independent experimental repeats.

    (3) The study lacks data on the cellular distribution of endoglin and THSD7A:

    a) Figure 6 - Is THSD7A expected to be present in the nucleus as shown in panel D (label D is missing in the Figure). It is not clear if this is observed in neurons. a Western of endogenous THSD7A on cell fractions would clarify this. The authors should further characterize the cellular distribution of THSD7A in both cell types. Similarly, the cellular distribution of endoglin in the cancer cells should be provided. This would help validate the proposed model in Figure 8.

    b) Figure 7 - Although the western blot provides convincing evidence for the role of endoglin in THSD7A trafficking, the microscopy data lack resolution as well as key analyses. While differences between shSCR and shEng cells are clear visually, the insets appear to be zoomed digitally which decreases resolution and interferes with interpretation. It would be crucial to show the colocalization of endoglin and THSD7A within CD63-postive MVE structures. What are the structures in Figure 7E shSCR zoom1? It would be important to rule out that these are migrasomes using TSPAN4 staining. More information on how the analysis was conducted is needed (i.e. how extracellular areas were chosen and whether the images are representative of the larger population). A widefield image of shSCR and shEng cells and DAPI or HOECHST staining in the higher magnification images should be provided. Additionally, the authors should quantify the colocalization of external CD63 and mScarlet signals from many independently acquired images (as they did for the internal signals in panel F). Is there no external THSD7A signal in the shEng cells?

  5. Author response:

    Public Reviews:

    Reviewer #1 (Public review):

    Summary:

    The study significantly advances our understanding of how exosomes regulate filopodia formation. Filopodia play crucial roles in cell movement, polarization, directional sensing, and neuronal synapse formation. McAtee et al. demonstrated that exosomes, particularly those enriched with the protein THSD7A, play a pivotal role in promoting filopodia formation through Cdc42 in cancer cells and neurons. This discovery unveils a new extracellular mechanism through which cells can control their cytoskeletal dynamics and interaction with their surroundings. The study employs a combination of rescue experiments, live-cell imaging, cell culture, and proteomic analyses to thoroughly investigate the role of exosomes and THSD7A in filopodia formation in cancer cells and neurons. These findings offer valuable insights into fundamental biological processes of cell movement and communication and have potential implications for understanding cancer metastasis and neuronal development.

    Weaknesses:

    The conclusions of this study are in most cases supported by data, but some aspects of data analysis need to be better clarified and elaborated. Some conclusions need to be better stated and according to the data observed.

    We appreciate the reviewer's recognition of the impact of our study. We will address the concerns about data analysis and statement of our conclusions in our full response to reviewers.

    Reviewer #2 (Public review):

    Summary:

    The authors show that small EVs trigger the formation of filopodia in both cancer cells and neurons. They go on to show that two cargo proteins, endoglin, and THSD7A, are important for this process. This possibly occurs by activating the Rho-family GTPase CDC42.

    Strengths:

    The EV work is quite strong and convincing. The proteomics work is well executed and carefully analyzed. I was particularly impressed with the chick metastasis assay that added strong evidence of in vivo relevance.

    Weaknesses:

    The weakest part of the paper is the Cdc42 work at the end of the paper. It is incomplete and not terribly convincing. This part of the paper needs to be improved significantly.

    We appreciate the reviewer's recognition of the impact of our study. Indeed, more work needs to be done to clarify the role of Cdc42 in the induction of filopodia by exosome-associated THSD7A. We anticipate that this will be a separate manuscript, delving in-depth into how exosome-associated THSD7A interacts with recipient cells to activate Cdc42 and carrying out a variety of assays for Cdc42 activation.

    Reviewer #3 (Public review):

    Summary:

    The authors identify a novel relationship between exosome secretion and filopodia formation in cancer cells and neurons. They observe that multivesicular endosomes (MVE)-plasma membrane (PM) fusion is associated with filopodia formation in HT1080 cells and that MVEs are present in filopodia in primary neurons. Using overexpression and knockdown (KD) of Rab27/HRS in HT1080 cells, melanoma cells, and/or primary rat neurons, they found that decreasing exosome secretion reduces filopodia formation, while Rab27 overexpression leads to the opposite result. Furthermore, the decreased filopodia formation is rescued in the Rab27a/HRS KD melanoma cells by the addition of small extracellular vesicles (EVs) but not large EVs purified from control cells. The authors identify endoglin as a protein unique to small EVs secreted by cancer cells when compared to large EVs. KD of endoglin reduces filopodia formation and this is rescued by the addition of small EVs from control cells and not by small EVs from endoglin KD cells. Based on the role of filopodia in cancer metastasis, the authors then investigate the role of endoglin in cancer cell metastasis using a chick embryo model. They find that injection of endoglin KD HT1080 cells into chick embryos gives rise to less metastasis compared to control cells - a phenotype that is rescued by the co-injection of small EVs from control cells. Using quantitative mass spectrometry analysis, they find that thrombospondin type 1 domain containing 7a protein (THSD7A) is downregulated in small EVs from endoglin KD melanoma cells compared to those from control cells. They also report that THSD7A is more abundant in endoglin KD cell lysate compared to control HT1080 cells and less abundant in small EVs from endoglin KD cells compared to control cells, indicating a trafficking defect. Indeed, using immunofluorescence microscopy, the authors observe THSD7A-mScarlet accumulation in CD63-positive structures in endoglin KD HT1080 cells, compared to control cells. Finally, the authors determine that exosome-secreted THSD7A induces filopodia formation in a Cdc42-dependent mechanism.

    Strengths:

    (1) While exosomes are known to play a role in cell migration and autocrine signaling, the relationship between exosome secretion and the formation of filopodia is novel.

    (2) The authors identify an exosomal cargo protein, THSD7A, which is essential for regulating this function.

    (3) The data presented provide strong evidence of a role for endoglin in the trafficking of THSD7A in exosomes.

    (4) The authors associate this process with functional significance in cancer cell metastasis and neurological synapse formation, both of which involve the formation of filopodia.

    (5) The data are presented clearly, and their interpretation appropriately explains the context and significance of the findings.

    Weaknesses:

    (1) A better characterization of the nature of the small EV population is missing:

    It is unclear why the authors chose to proceed to quantitative mass spectrometry with the bands in the Coomassie from size-separated EV samples, as there are other bands present in the small EV lane but not the large EV lane. This is important to clarify because it underlies how they were able to identify THSD7A as a unique regulator of exosome-mediated filopodia formation. Is there a reason why the total sample fractions were not compared? This would provide valuable information on the nature of the small and large EV populations.

    We would like to clarify that there are two sets of proteomics data in the manuscript. The first was comparing bands from a Coomassie gel from two samples: small EVs and large EVs from B16F1 cells. In this proteomics experiment, we identified endoglin as present in small EVs, but not large EVs. For this experiment, we only sent 4 bands from the small EV lane, chosen based on their obvious banding pattern difference on the Coomassie gel.

    In the second proteomics experiment, we used quantitative iTRAQ proteomics to compare small EVs purified from B16F1 control (shScr) and endoglin KD (shEng1 and shEng2) cell lines. In this experiment, we sent total protein extracted from small EV samples for analysis. So, these samples included the entire EV content, not just selected bands from a gel. In this experiment, we identified THSD7A as reduced in the shEng small EVs.

    (2) Data analysis and quantification should be performed with increased rigor:

    a) Figure 1C - The optical and temporal resolution are insufficient to conclusively characterize the association between exosome secretion and filopodia. Specifically, the 10-second interval used in the image acquisitions is too close to the reported 20-second median time between exosome secretion and filopodia formation. Two-5 sec intervals should be used to validate this. It would also be important to correlate the percentage of filopodia events that co-occur with exosome secretion. Is this a phenomenon that occurs with most or only a small number of filopodia? Additionally, resolution with typical confocal microscopy is subpar for these analyses. TIRF microscopy would offer increased resolution to parse out secretion events. As the TIRF objective is listed in the Methods section, figure legends should mention which images were acquired using TIRF microscopy.

    We acknowledge that the frame rate naturally limits our estimates of the timing of filopodia formation after exosome secretion. We set out to show a relationship between exosome secretion and filopodia formation, based on their proximity in timing. While our data set shows a median time interval of 20 seconds, the true median could be between 10-30 seconds, based on our frame rate. Regardless of the exact timing, our data show that exosome secretion is rapidly followed by filopodia formation events.

    To address the question of the percentage of filopodia events that are preceded by exosome secretion, the reviewer is correct in stating that we might need TIRF microscopy to get an accurate calculation of this number. Nonetheless, we will review our live imaging data for this experiment to determine if this calculation is possible. Again, we will be limited by the frame rate we used to capture the images, so we could possibly be missing secretion events taking place between the 10 second time intervals. Regardless, for the secretion events that we visualized, we always observed subsequent filopodia formation.

    No TIRF imaging was used in this manuscript. A TIRF objective was used for selected neuron imaging (see methods); however, it was used for spinning disk confocal microscopy, not for TIRF imaging. We will clarify this in the methods.

    b) Figure 2 - It would be important to perform further analysis to concretely determine the relationship between exosome secretion and filopodia stability. Are secretion events correlated with the stability of filopodia? Is there a positive feedback loop that causes further filopodia stability and length with increased secretion? Furthermore, is there an association between the proximity of secretion with stability? Quantification of filopodia more objectively (# of filopodia/cell) would be helpful.

    Our data shows that manipulation of general exosome secretion, via Hrs knockdown, affects both de novo filopodia formation and filopodia stability (Fig 2g,h). Interestingly, knockdown of endoglin only affects de novo filopodia formation, while filopodia stability is unaffected (Fig 4g,h). These results suggest that filopodia stability is dependent upon exosome cargoes besides endoglin/THSD7A. Such cargoes might include other extracellular matrix molecules, such as fibronectin. We previously showed that exosomes promote nascent cell adhesion and rapid cell migration, through exosome-bound fibronectin (Sung et al., Nature Communications, 6:7164, 2015). We also previously found that inhibition of exosome secretion affects the persistence of invadopodia, which are filopodia-dependent structures (Hoshino et al., Cell Reports, 5:1159-1168, 2013). We agree that this is an interesting research direction, and perhaps future work could focus on exosomal factors that are responsible for filopodia persistence.

    With regard to the way we plotted the filopodia data, we plotted the cancer cell data as filopodia per cell area so that it matched the neuron data, which was plotted as filopodia per 100 mm of dendrite distance. Since the neurons cannot be imaged as a whole cell, the quantification is based on the length of the dendrite in the image. We found that graphing the cancer cell data as filopodia per cell gave similar results as filopodia per cell area, as there were no significant differences in cell area between conditions and experiments. We plan to include a new supplementary figure showing the data in Figure 2 plotted as filopodia per cell to show that this quantification gives the same results.

    c) Figure 6 - Why use different gel conditions to detect THSD7A in small EVs from B16F1 cells vs HT1080 and neurons? Why are there two bands for THSD7A in panels C and E? It is difficult to appreciate the KD efficiency in E. The absence of a signal for THSD7A in the HT1080 shEng small EVs that show a signal for endoglin is surprising. The authors should provide rigorous quantification of the westerns from several independent experimental repeats.

    Detection of THSD7A via Western blot was, unfortunately, not straightforward and simple. Due to the large size (~260 kDa) of THSD7A, its low level of expression in cancer cells, as well as the inconsistency of commercially available THSD7A antibodies, we had to troubleshoot multiple conditions. We found that it was much easier to detect THSD7A in the human fibrosarcoma cell line HT1080 than in the mouse B16F1 cells, both in the cell lysates and in the small EVs. We were usually unable to detect THSD7A using these same conditions for the mouse melanoma B16F1 samples, but were successful using native gel conditions. We also detected THSD7A in rat primary neuron samples. All these samples were from different source organisms (human, mouse, rat) and from either cell lysates or extracellular vesicles, further complicating the analyses. Expression and maturation of THSD7A in these different cell types and compartments could involve different post-translational modifications, such as glycosylation, thus requiring different methods needed to detect THSD7A on Western blots and leading to different banding patterns. Based on our THSD7A trafficking data, we believe that in control cells, most of the THSD7A is getting trafficked and secreted via small EVs. As you can see in Figure 7A, the band for THSD7A in the shScr cell lysate is relatively light and also shows a double band similar to Figure 6E (both HT1080 samples).

    With regard to the level of knockdown of THSD7A in the Western blot shown in Figure 6E, the normalized level is quantitated below the bands. If you compare that quantitation to the filopodia phenotypes in the same panel, they are quite concordant. Figures 7B and 7C show quantification of triplicate Western blots, highlighting the significant accumulation of THSD7A in shEng cell lysates, as well as significant small EV secretion of THSD7A in control and WT rescued conditions.

    (3) The study lacks data on the cellular distribution of endoglin and THSD7A:

    a) Figure 6 - Is THSD7A expected to be present in the nucleus as shown in panel D (label D is missing in the Figure). It is not clear if this is observed in neurons. a Western of endogenous THSD7A on cell fractions would clarify this. The authors should further characterize the cellular distribution of THSD7A in both cell types. Similarly, the cellular distribution of endoglin in the cancer cells should be provided. This would help validate the proposed model in Figure 8.

    The image in figure 6D shows an HT1080 cell stained with phalloidin-Alexa Fluor 488 to visualize F-actin with or without expression of THSD7A-mScarlet. In order to fully visualize the thin filopodia protrusions, the cellular plane of focus of the images for this panel was purposely taken at the bottom of the cell, where the cell is attached to the coverslip glass. Thus, we interpret the red signal across the cell body as THSD7A-mScarlet expression on the plasma membrane underneath the cell, not in the nucleus. The neuron images only include the dendrite portion of the neurons; therefore, there is no nucleus present in the neuronal images.

    b) Figure 7 - Although the western blot provides convincing evidence for the role of endoglin in THSD7A trafficking, the microscopy data lack resolution as well as key analyses. While differences between shSCR and shEng cells are clear visually, the insets appear to be zoomed digitally which decreases resolution and interferes with interpretation. It would be crucial to show the colocalization of endoglin and THSD7A within CD63-postive MVE structures. What are the structures in Figure 7E shSCR zoom1? It would be important to rule out that these are migrasomes using TSPAN4 staining. More information on how the analysis was conducted is needed (i.e. how extracellular areas were chosen and whether the images are representative of the larger population). A widefield image of shSCR and shEng cells and DAPI or HOECHST staining in the higher magnification images should be provided. Additionally, the authors should quantify the colocalization of external CD63 and mScarlet signals from many independently acquired images (as they did for the internal signals in panel F). Is there no external THSD7A signal in the shEng cells?

    The images for Figure 7E were taken with high resolution on a confocal microscope. Insets for Figure 7E were zoomed in so that readers could see the tiny structures. Zoom 1 in Figure 7E shows areas of extracellular deposition. In these areas, we can see small punctate depositions that are positive for CD63 and/or THSD7A-mScarlet. Our interpretation of this staining is that the cells are secreting heterogeneous small EVs that are then attached to the glass coverslip. The images and zooms in Fig 7E were chosen to be representative and indeed reveal that there is more extracellular deposition of THSD7A-mScarlet outside the control shScr cells compared to the shEng cells, consistent with more export of THSD7A into small EVs from shScr cells when compared to those of shEng cells (Fig 7A,B). However, we did not quantify this difference, as these experiments were conducted with transient transfection of THSD7A-mScarlet and it is challenging to determine which cell the extracellular THSD7A-mScarlet came from, complicating any quantitative analysis on a per-cell basis. Quantification of internal THSD7A localization is much more straightforward in this experimental regime. Indeed, in Figure 7F we assessed internal colocalization of THSD7A-mScarlet and CD63, which we obtained by choosing only cells that were visually positive for THSD7A-mScarlet in each transient transfection and omitting all extracellular signals. Quantifying the extracellular colocalization of THSD7A and CD63 could certainly be a future direction for this project and would require establishing cells that stably express THSD7A-mScarlet.