(Translated by https://www.hiragana.jp/)
Invadopodium Assembly - MBInfo Wiki
The Wayback Machine - https://web.archive.org/web/20160323161643/http://www.mechanobio.info/topics/cytoskeleton-dynamics/go-0071437/Invadopodium-Assembly

Invadopodium Assembly


Initiation of invadopodium assembly[Edit]

Initiation of invadopdia formation is highly complex, being influenced by various signalling cascades and phosphorylation events that occur following detection of a stimulant. 

For example, activation of the epidermal growth factor (EGF) receptor by EGF has been shown to trigger and/or enhance invadopodia formation [1] as have platelet-derived growth factor (PDGF) and reactive oxygen species (ROS) (as reviewed in [2]). Following stimulation of the EGF receptor, the tyrosine kinase Src is activated, followed by the Abelson-related nonreceptor tyrosine kinase, Arg [3]. Src has traditionally been known as a vital component in the signaling cascade that governs the initiation of invadopodia formation [4, 5, 6, 7] however more recent studies have also highlighted the importance of other Src family kinases [8] and protein kinase C [9] in this process.

Activation of these kinases are reported to increase the number of free actin barbed ends and lead to the phosphorylation of cortactin [10] and the scaffolding protein Tks5 (tyrosine kinase substrate with 5 SH3 domains) [7]. Together these events promote actin assembly polymerization and maturation of the invadopodium [10]. Tks5 is also associated with the production of invadopodia in response to ROS [11].

In addition, the GTPase Cdc42 has been implicated in invadopodia formation via the regulation of neuronal Wiskott-Aldrich Syndrome protein (N-WASP), which acts in concert with the Arp2/3 complex to promote actin polymerization [9]. Similarly, cortactin is able to promote actin polymerization through binding and activating the Arp2/3 complex [12]. Live-cell imaging experiments have shown the accumulation of cortactin in invadopodia precedes matrix metalloproteinases (MMPs) accumulation and matrix degradation [5].

Ultimately the outcome of initiating invadopodia formation has been speculated to result in the production of a podosome-like precursor. This precursor structure comprises a small branched actin network, which is suggested to continue to polymerize and extend deeper into the extracellular matrix (ECM) [13] as MMPs are released from the tip of the elongating structure.

Extension of the Invadopodia[Edit]

Following initiation by an appropriate signal, the actin cytoskeleton is reorganized to facilitate invadopodia formation. Although several kinases and GTPases are known to influence the transduction of extracellular signaling events leading to F-actin nucleation, there is currently no clear consensus on the precise steps involved [14]. The phosphorylation of cortactin is known occur prior to F-actin nucleation [8], after which arrays of actin filaments begin to assemble. 
 
Two distinct types of filamentous actin networks are suggested to cooperatively form invadopodia - a branched actin network that forms the base of the structure and a parallel array of bundled actin filaments that form along the length of the invadopodial shaft. The latter is responsible for its elongation [15] and works together with microtubules and intermediate filaments to extend the invadopodia [15].

The production of these two actin networks are driven by different molecular components. Formation of the branched array of actin filaments at the base of invadopodia is driven by Arp2/3 and its activator N-WASP [15, 16, 1]. The activation of N-WASP is facilitated by the Rho GTPase Cdc42, WIP (WASP interacting protein) and Nck1 (non-catalytic region of tyrosine kinase adaptor protein 1) - all of which have been found to localize to invadopodia and are necessary for their formation [1]. The resulting structure resembles the actin core of podosomes.

Elongation of invadopodia is suggested to be driven by the extension of parallel arrays of bundled actin filaments that are present along the length of invadopodia [15]. Extension of these filaments is driven by formins (Functional Module: Formin and Profilin in Actin Nucleation), whilst their bundling is coordinated by the actin cross-linker fascin. Studies have shown both formins [15, 17] and fascin [18] localize to invadopodia and are necessary for their formation and stability. Additional proteins required for maintaining the stability of these structures include members of the Ena/VASP family. The invasive isoform of the Ena/VASP protein MenaINV has been shown to stabilize invadopodia and is hypothesized to do so either via stimulating actin polymerization from free barbed ends or through the bundling of actin filaments [19].

Invadopodia extension is driven by actin polymerization and hence the concentration of G-actin-ATP monomers at the invadopodia tip contributes to the rate of extension. The actin severing protein cofilin has been shown to generate free barbed ends onto which G-actin monomers can be added [20]. The activity of cofilin is focused within the core of the invadopodium by the action of the RhoGTPase RhoC [21]. RhoC activity is regulated by its activator, the guanine nucleotide exchange factor (GEF) p190RhoGEF and its deactivator, the GTPase activating protein (GAP) p190RhoGAP. The phosphorylation of cofilin by RhoC decreases its severing ability, therefore in order to promote extension of the invadopodium, RhoC activity is limited to regions beyond the invadopodium core. This is achieved by restricting the localization of the RhoC activator p190RhoGEF to regions surrounding invadopodia, whilst the RhoC deactivator p190RhoGAP localizes within invadopodia [21]. The spatially regulated activity of RhoC, and subsequently cofilin, leads to barbed end production within invadopodia, which facilitates actin polymerization and filament extension.

The rate of extension is also dependent upon the ability of the growing filaments to overcome membrane resistance and the concomitant incorporation of new membrane. This can be achieved through the addition of membrane from vesicles delivered to sites of invadopodia formation. The Golgi apparatus is found in close proximity to invadopodia and is speculated to be positioned in such a manner to meet the demands of new membrane incorporation [22]. New membrane must be remodeled to allow for the growth of invadopodia into the extracellular matrix (ECM). F-BAR proteins have long been known to modulate membrane curvature and recently the F-BAR protein, CIP4 (Cdc42 interacting protein 4), has been implicated in invadopodia as both a scaffold and a means to promote membrane curvature [23]. The combination of actin polymerization and membrane incorporation ultimately results in the extension of invadopodia into the extracellular matrix (ECM).

Degradation of the extra-cellular matrix[Edit]

The main function attributed to invadopodia is that of extracellular matrix (ECM) degradation, facilitated by the secretion of proteases. Maintenance of this process requires the delivery of new proteases from the Golgi, which is conveniently positioned in close proximity to invadopodia [22]. Indeed, the treatment of cells with Brefeldin A, an inhibitor of ER (endoplasmic reticulum) to Golgi transport, has been shown to prevent ECM degradation by invadopodia [14]. The Ena/VASP family protein, Mena, is also implicated in this process. More specifically the MenaINV isoform that favors cancer cell invasiveness by promoting the stabilization of invadopodia and enhancing their ECM-degrading activity [24, 25]. 

ECM degradation itself is carried out by a variety of secreted matrix metalloproteinases (MMPs) and serine proteases. Currently over 25 different MMPs have been identified that together have the potential to degrade the entire ECM [26]. Of all the MMPs, MMP14 (also known as MT1-MMP), is considered to be the major regulator of invadopodia-mediated ECM degradation across several cell models [27]. MMP docking has been shown to be essential for ECM degradation in melanoma cells, with knock-down [5] or overexpression [26] showing a decrease or increase in invadopodial activity, respectively. Cortactin, commonly known as a weak activator of the actin nucleator Arp2/3, also has a role to play in MMP secretion and ECM degradation [27].

Cortactin accumulation has been shown to precede MMP accumulation at the tips of invadopodia and has been suggested to regulate their secretion [5]. This cortactin-dependent maturation process has been shown to be dependent on the activity of LIM kinases [28].

Invadopodia disassembly[Edit]

The final step in function of invadopodia is disassembly which primarily involves dismantling the actin core [29]. Several proteins have been implicated in a cascade leading to this, including paxillin, extracellular signal-regulated kinases (Erk) and calpain [29]. The phosphorylation state of tyrosine residues within paxillin localized to invadopodia controls the rate of disassembly. The mutation of specific tyrosine residues in paxillin render these sites non-phosphorylatable and this results in a significant delay in the disassembly of invadopodia. Furthermore, phosphorylation of paxillin was shown to promote the activation of Erk, which promotes the activation of calpain [29]. Calpains are calcium dependent, non-lysosomal, cysteine proteases. The activation of calpain 2 has been shown to be required for the degradation of cortactin, a component of the actin core and this subsequently promotes invadopodia disassembly [30]. The inhibition of calpain [29] or the expression of calpain-resistant cortactin in cells lacking endogenous cortactin [30], results in a decrease in the rate of invadopodia disassembly. This is akin to the role of calpain in the degradation of talin [31] and focal adhesion kinase [32], which has been shown to promote focal adhesion disassembly.

References

  1. Yamaguchi H., Lorenz M., Kempiak S., Sarmiento C., Coniglio S., Symons M., Segall J., Eddy R., Miki H., Takenawa T., Condeelis J. Molecular mechanisms of invadopodium formation: the role of the N-WASP-Arp2/3 complex pathway and cofilin. J. Cell Biol. 2005; 168(3). [PMID: 15684033]
  2. Murphy DA., Courtneidge SA. The 'ins' and 'outs' of podosomes and invadopodia: characteristics, formation and function. Nat. Rev. Mol. Cell Biol. 2011; 12(7). [PMID: 21697900]
  3. Kimura F., Iwaya K., Kawaguchi T., Kaise H., Yamada K., Mukai K., Matsubara O., Ikeda N., Kohno N. Epidermal growth factor-dependent enhancement of invasiveness of squamous cell carcinoma of the breast. Cancer Sci. 2010; 101(5). [PMID: 20219074]
  4. Tarone G., Cirillo D., Giancotti FG., Comoglio PM., Marchisio PC. Rous sarcoma virus-transformed fibroblasts adhere primarily at discrete protrusions of the ventral membrane called podosomes. Exp. Cell Res. 1985; 159(1). [PMID: 2411576]
  5. Artym VV., Zhang Y., Seillier-Moiseiwitsch F., Yamada KM., Mueller SC. Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006; 66(6). [PMID: 16540652]
  6. Kelley LC., Ammer AG., Hayes KE., Martin KH., Machida K., Jia L., Mayer BJ., Weed SA. Oncogenic Src requires a wild-type counterpart to regulate invadopodia maturation. J. Cell. Sci. 2010; 123(Pt 22). [PMID: 20980387]
  7. Stylli SS., Stacey TT., Verhagen AM., Xu SS., Pass I., Courtneidge SA., Lock P. Nck adaptor proteins link Tks5 to invadopodia actin regulation and ECM degradation. J. Cell. Sci. 2009; 122(Pt 15). [PMID: 19596797]
  8. Ayala I., Baldassarre M., Giacchetti G., Caldieri G., Tetè S., Luini A., Buccione R. Multiple regulatory inputs converge on cortactin to control invadopodia biogenesis and extracellular matrix degradation. J. Cell. Sci. 2008; 121(Pt 3). [PMID: 18198194]
  9. Mader CC., Oser M., Magalhaes MA., Bravo-Cordero JJ., Condeelis J., Koleske AJ., Gil-Henn H. An EGFR-Src-Arg-cortactin pathway mediates functional maturation of invadopodia and breast cancer cell invasion. Cancer Res. 2011; 71(5). [PMID: 21257711]
  10. Tehrani S., Tomasevic N., Weed S., Sakowicz R., Cooper JA. Src phosphorylation of cortactin enhances actin assembly. Proc. Natl. Acad. Sci. U.S.A. 2007; 104(29). [PMID: 17606906]
  11. Diaz B., Shani G., Pass I., Anderson D., Quintavalle M., Courtneidge SA. Tks5-dependent, nox-mediated generation of reactive oxygen species is necessary for invadopodia formation. Sci Signal 2009; 2(88). [PMID: 19755709]
  12. Uruno T., Liu J., Zhang P., Fan Yx undefined., Egile C., Li R., Mueller SC., Zhan X. Activation of Arp2/3 complex-mediated actin polymerization by cortactin. Nat. Cell Biol. 2001; 3(3). [PMID: 11231575]
  13. The matrix corroded: podosomes and invadopodia in extracellular matrix degradation. Trends Cell Biol. 2007; 17(3). [PMID: 17275303]
  14. Buccione R., Caldieri G., Ayala I. Invadopodia: specialized tumor cell structures for the focal degradation of the extracellular matrix. Cancer Metastasis Rev. 2009; 28(1-2). [PMID: 19153671]
  15. Schoumacher M., Goldman RD., Louvard D., Vignjevic DM. Actin, microtubules, and vimentin intermediate filaments cooperate for elongation of invadopodia. J. Cell Biol. 2010; 189(3). [PMID: 20421424]
  16. Lorenz M., Yamaguchi H., Wang Y., Singer RH., Condeelis J. Imaging sites of N-wasp activity in lamellipodia and invadopodia of carcinoma cells. Curr. Biol. 2004; 14(8). [PMID: 15084285]
  17. Lizárraga F., Poincloux R., Romao M., Montagnac G., Le Dez G., Bonne I., Rigaill G., Raposo G., Chavrier P. Diaphanous-related formins are required for invadopodia formation and invasion of breast tumor cells. Cancer Res. 2009; 69(7). [PMID: 19276357]
  18. Li A., Dawson JC., Forero-Vargas M., Spence HJ., Yu X., König I., Anderson K., Machesky LM. The actin-bundling protein fascin stabilizes actin in invadopodia and potentiates protrusive invasion. Curr. Biol. 2010; 20(4). [PMID: 20137952]
  19. Hori T., Ito F. [Prostaglandin production by growth factors and its biological activity]. Tanpakushitsu Kakusan Koso 1991; 36(7). [PMID: 1908107]
  20. Oser M., Yamaguchi H., Mader CC., Bravo-Cordero JJ., Arias M., Chen X., Desmarais V., van Rheenen J., Koleske AJ., Condeelis J. Cortactin regulates cofilin and N-WASp activities to control the stages of invadopodium assembly and maturation. J. Cell Biol. 2009; 186(4). [PMID: 19704022]
  21. Bravo-Cordero JJ., Oser M., Chen X., Eddy R., Hodgson L., Condeelis J. A novel spatiotemporal RhoC activation pathway locally regulates cofilin activity at invadopodia. Curr. Biol. 2011; 21(8). [PMID: 21474314]
  22. Baldassarre M., Pompeo A., Beznoussenko G., Castaldi C., Cortellino S., McNiven MA., Luini A., Buccione R. Dynamin participates in focal extracellular matrix degradation by invasive cells. Mol. Biol. Cell 2003; 14(3). [PMID: 12631724]
  23. Pichot CS., Arvanitis C., Hartig SM., Jensen SA., Bechill J., Marzouk S., Yu J., Frost JA., Corey SJ. Cdc42-interacting protein 4 promotes breast cancer cell invasion and formation of invadopodia through activation of N-WASp. Cancer Res. 2010; 70(21). [PMID: 20940394]
  24. Albiges-Rizo C., Destaing O., Fourcade B., Planus E., Block MR. Actin machinery and mechanosensitivity in invadopodia, podosomes and focal adhesions. J. Cell. Sci. 2009; 122(Pt 17). [PMID: 19692590]
  25. Philippar U., Roussos ET., Oser M., Yamaguchi H., Kim HD., Giampieri S., Wang Y., Goswami S., Wyckoff JB., Lauffenburger DA., Sahai E., Condeelis JS., Gertler FB. A Mena invasion isoform potentiates EGF-induced carcinoma cell invasion and metastasis. Dev. Cell 2008; 15(6). [PMID: 19081071]
  26. Nakahara H., Howard L., Thompson EW., Sato H., Seiki M., Yeh Y., Chen WT. Transmembrane/cytoplasmic domain-mediated membrane type 1-matrix metalloprotease docking to invadopodia is required for cell invasion. Proc. Natl. Acad. Sci. U.S.A. 1997; 94(15). [PMID: 9223295]
  27. Clark ES., Whigham AS., Yarbrough WG., Weaver AM. Cortactin is an essential regulator of matrix metalloproteinase secretion and extracellular matrix degradation in invadopodia. Cancer Res. 2007; 67(9). [PMID: 17483334]
  28. Scott RW., Hooper S., Crighton D., Li A., König I., Munro J., Trivier E., Wickman G., Morin P., Croft DR., Dawson J., Machesky L., Anderson KI., Sahai EA., Olson MF. LIM kinases are required for invasive path generation by tumor and tumor-associated stromal cells. J. Cell Biol. 2010; 191(1). [PMID: 20876278]
  29. Badowski C., Pawlak G., Grichine A., Chabadel A., Oddou C., Jurdic P., Pfaff M., Albigès-Rizo C., Block MR. Paxillin phosphorylation controls invadopodia/podosomes spatiotemporal organization. Mol. Biol. Cell 2008; 19(2). [PMID: 18045996]
  30. McMaster OG., Du F., French ED., Schwarcz R. Focal injection of aminooxyacetic acid produces seizures and lesions in rat hippocampus: evidence for mediation by NMDA receptors. Exp. Neurol. 1991; 113(3). [PMID: 1833221]
  31. Franco SJ., Rodgers MA., Perrin BJ., Han J., Bennin DA., Critchley DR., Huttenlocher A. Calpain-mediated proteolysis of talin regulates adhesion dynamics. Nat. Cell Biol. 2004; 6(10). [PMID: 15448700]
  32. Chan KT., Bennin DA., Huttenlocher A. Regulation of adhesion dynamics by calpain-mediated proteolysis of focal adhesion kinase (FAK). J. Biol. Chem. 2010; 285(15). [PMID: 20150423]
Updated on: Tue, 28 Oct 2014 14:53:03 GMT