Invadopodia

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 360 Experts worldwide ranked by ideXlab platform

Hideki Yamaguchi - One of the best experts on this subject based on the ideXlab platform.

  • cdcp1 regulates the function of mt1 mmp and Invadopodia mediated invasion of cancer cells
    Molecular Cancer Research, 2013
    Co-Authors: Yuri Miyazawa, Hideki Yamaguchi, Motoharu Seiki, Takamasa Uekita, Yuumi Ito, Ryuichi Sakai
    Abstract:

    Complement C1r/C1s, Uegf, Bmp1 (CUB) domain-containing protein 1 (CDCP1) is a transmembrane protein that regulates anchorage-independent growth and cancer cell migration and invasion. Expression of CDCP1 is detected in a number of cancer cell lines and tissues and is closely correlated with poor prognosis. Invadopodia are actin-based protrusions on the surface of invasive cancer cells that promote the degradation of the extracellular matrix (ECM) via localized proteolysis, which is mainly mediated by membrane type 1 matrix metalloproteinase (MT1-MMP). MT1-MMP is accumulated at Invadopodia by targeted delivery via membrane trafficking. The present study shows that CDCP1 is required for ECM degradation by Invadopodia in human breast cancer and melanoma cells. CDCP1 localized to caveolin-1-containing vesicular structures and lipid rafts and was detected in close proximity to Invadopodia. Further biochemical analysis revealed that substantial amounts of CDCP1 existed in the Triton X-100 insoluble lipid raft fraction. CDCP1 was coimmunoprecipitated with MT1-MMP and colocalized with MT1-MMP at the vesicular structures. The siRNA-mediated knockdown of the CDCP1 expression markedly inhibited MT1-MMP-dependent ECM degradation and Matrigel invasion and reduced the accumulation of MT1-MMP at Invadopodia, as shown by immunofluorescence analysis. These results indicate that CDCP1 is an essential regulator of the trafficking and function of MT1-MMP- and Invadopodia-mediated invasion of cancer cells.

  • Pathological roles of Invadopodia in cancer invasion and metastasis.
    European journal of cell biology, 2012
    Co-Authors: Hideki Yamaguchi
    Abstract:

    Invadopodia are actin-rich membrane protrusions formed by invasive cancer cells. Invadopodia mediate the focal degradation of pericellular extracellular matrix (ECM) by the localized proteolytic activity of matrix metalloproteinases (MMPs). Over the last 2 decades, much progress has been made in identifying the molecular components of Invadopodia and understanding the molecular mechanisms underlying their formation. Although the physiological and pathological roles of Invadopodia have long been elusive, emerging evidence has begun to reveal their importance in local invasion during cancer metastasis. This review highlights recent findings on the roles of Invadopodia in cancer invasion and metastasis and discusses the possibility of and strategies for targeting Invadopodia formation for the development of novel anticancer therapeutics.

  • N-WASP-mediated invadopodium formation is involved in intravasation and lung metastasis of mammary tumors.
    Journal of cell science, 2012
    Co-Authors: Bojana Gligorijevic, Hideki Yamaguchi, Jeffrey Wyckoff, Yarong Wang, Evanthia T Roussos, John Condeelis
    Abstract:

    Invadopodia are proteolytic membrane protrusions formed by highly invasive cancer cells, commonly observed on substrate(s) mimicking extracellular matrix. Although Invadopodia are proposed to have roles in cancer invasion and metastasis, direct evidence has not been available. We previously reported that neural Wiskott-Aldrich syndrome protein (N-WASP), a member of WASP family proteins that regulate reorganization of the actin cytoskeleton, is an essential component of Invadopodia. Here, we report that N-WASP-mediated invadopodium formation is essential in breast cancer invasion, intravasation and lung metastasis. We established stable cell lines based on MTLn3 rat mammary adenocarcinoma cells that either overexpressed a dominant-negative (DN) N-WASP construct or in which N-WASP expression was silenced by a pSuper N-WASP shRNA. Both the N-WASP shRNA and DN N-WASP cells showed a markedly decreased ability to form Invadopodia and degrade extracellular matrix. In addition, formation of Invadopodia in primary tumors and collagen I degradation were reduced in the areas of invasion (collagen-rich areas in the invasive edge of the tumor) and in the areas of intravasation (blood-vessel-rich areas). Our results suggest that tumor cells in vivo that have a decreased activity of N-WASP also have a reduced ability to form Invadopodia, migrate, invade, intravasate and disseminate to lung compared with tumor cells with parental N-WASP levels.

  • phosphoinositide 3 kinase signaling pathway mediated by p110α regulates Invadopodia formation
    Journal of Cell Biology, 2011
    Co-Authors: Hideki Yamaguchi, Shuhei Yoshida, Emi Muroi, Nachi Yoshida, Masahiro Kawamura, Zen Kouchi, Yoshikazu Nakamura, Ryuichi Sakai, Kiyoko Fukami
    Abstract:

    Invadopodia are extracellular matrix–degrading protrusions formed by invasive cancer cells that are thought to function in cancer invasion. Although many Invadopodia components have been identified, signaling pathways that link extracellular stimuli to Invadopodia formation remain largely unknown. We investigate the role of phosphoinositide 3-kinase (PI3K) signaling during Invadopodia formation. We find that in human breast cancer cells, both Invadopodia formation and degradation of a gelatin matrix were blocked by treatment with PI3K inhibitors or sequestration of D-3 phosphoinositides. Functional analyses revealed that among the PI3K family proteins, the class I PI3K catalytic subunit p110α, a frequently mutated gene product in human cancers, was selectively involved in Invadopodia formation. The expression of p110α with cancerous mutations promoted Invadopodia-mediated invasive activity. Furthermore, knockdown or inhibition of PDK1 and Akt, downstream effectors of PI3K signaling, suppressed Invadopodia formation induced by p110α mutants. These data suggest that PI3K signaling via p110α regulates Invadopodia-mediated invasion of breast cancer cells.

  • lipid rafts and caveolin 1 are required for Invadopodia formation and extracellular matrix degradation by human breast cancer cells
    Cancer Research, 2009
    Co-Authors: Hideki Yamaguchi, Shuhei Yoshida, Zen Kouchi, Yoshikazu Nakamura, Yukiko Takeo, Kiyoko Fukami
    Abstract:

    Invadopodia are ventral membrane protrusions through which invasive cancer cells degrade the extracellular matrix. They are thought to function in the migration of cancer cells through tissue barriers, which is necessary for cancer invasion and metastasis. Although many protein components of Invadopodia have been identified, the organization and the role of membrane lipids in Invadopodia are not well understood. In this study, the role of lipid rafts, which are cholesterol-enriched membrane microdomains, in the assembly and function of Invadopodia in human breast cancer cells was investigated. Lipid rafts are enriched, internalized, and dynamically trafficked at Invadopodia sites. Perturbation of lipid raft formation due to depleting or sequestering membrane cholesterol blocked the Invadopodia-mediated degradation of the gelatin matrix. Caveolin-1 (Cav-1), a resident protein of lipid rafts and caveolae, accumulates at Invadopodia and colocalizes with the internalized lipid raft membranes. Membrane type 1 matrix metalloproteinase (MT1-MMP), a matrix proteinase associated with Invadopodia, is localized at lipid raft-enriched membrane fractions and cotrafficked and colocalized with Cav-1 at Invadopodia. The small interfering RNA–mediated silencing of Cav-1 inhibited the Invadopodia-mediated and MT1-MMP–dependent degradation of the gelatin matrix. Furthermore, Cav-1 and MT1-MMP are coexpressed in invasive human breast cancer cell lines that have an ability to form Invadopodia. These results indicate that Invadopodia are the sites where enrichment and trafficking of lipid rafts occur and that Cav-1 is an essential regulator of MT1-MMP function and Invadopodia-mediated breast cancer cell invasion. [Cancer Res 2009;69(22):8594–602]

Susette C Mueller - One of the best experts on this subject based on the ideXlab platform.

  • dense fibrillar collagen is a potent inducer of Invadopodia via a specific signaling network
    Journal of Cell Biology, 2015
    Co-Authors: Vira V. Artym, Stephen Swatkoski, Kazue Matsumoto, Catherine Campbell, Ryan J Petrie, Emilios K Dimitriadis, Xin Li, Susette C Mueller, Thomas H Bugge
    Abstract:

    Cell interactions with the extracellular matrix (ECM) can regulate multiple cellular activities and the matrix itself in dynamic, bidirectional processes. One such process is local proteolytic modification of the ECM. Invadopodia of tumor cells are actin-rich proteolytic protrusions that locally degrade matrix molecules and mediate invasion. We report that a novel high-density fibrillar collagen (HDFC) matrix is a potent inducer of Invadopodia, both in carcinoma cell lines and in primary human fibroblasts. In carcinoma cells, HDFC matrix induced formation of Invadopodia via a specific integrin signaling pathway that did not require growth factors or even altered gene and protein expression. In contrast, phosphoproteomics identified major changes in a complex phosphosignaling network with kindlin2 serine phosphorylation as a key regulatory element. This kindlin2-dependent signal transduction network was required for efficient induction of Invadopodia on dense fibrillar collagen and for local degradation of collagen. This novel phosphosignaling mechanism regulates cell surface Invadopodia via kindlin2 for local proteolytic remodeling of the ECM.

  • dynamic membrane remodeling at Invadopodia differentiates Invadopodia from podosomes
    European Journal of Cell Biology, 2011
    Co-Authors: Vira V. Artym, Kazue Matsumoto, Susette C Mueller, Kenneth M Yamada
    Abstract:

    Invadopodia are specialized actin-rich protrusions of metastatic tumor and transformed cells with crucial functions in ECM degradation and invasion. Although early electron microscopy studies described Invadopodia as long filament-like protrusions of the cell membrane adherent to the matrix, fluorescence microscopy studies have focused on Invadopodia as actin-cortactin aggregates localized to areas of ECM degradation. The absence of a clear conceptual integration of these two descriptions of Invadopodial structure has impeded understanding of the regulatory mechanisms that govern Invadopodia. To determine the relationship between the membrane filaments identified by electron microscopy and the actin-cortactin aggregates of Invadopodia, we applied rapid live-cell high-resolution TIRF microscopy to examine cell membrane dynamics at the cortactin core of the Invadopodia of human carcinoma cells. We found that cortactin docking to the cell membrane adherent to 2D fibronectin matrix initiates invadopodium assembly associated with the formation of an Invadopodial membrane process that extends from a ventral cell membrane lacuna toward the ECM. The tip of the Invadopodial process flattens as it interacts with the 2D matrix, and it undergoes constant rapid ruffling and dynamic formation of filament-like protrusions as the invadopodium matures. To describe this newly discovered dynamic relationship between the actin-cortactin core and Invadopodial membranes, we propose a model of the Invadopodial complex. Using TIRF microscopy, we also established that - in striking contrast to the invadopodium - membrane at the podosome of a macrophage fails to form any process- or filament-like membrane protrusions. Thus, the undulation and ruffling of the Invadopodial membrane together with the formation of dynamic filament-like extensions from the Invadopodial cortactin core defines Invadopodia as invasive superstructures that are distinct from the podosomes.

  • c src differentially regulates the functions of microtentacles and Invadopodia
    Oncogene, 2010
    Co-Authors: Eric M Balzer, Susette C Mueller, Toshiyuki Yoneda, Rebecca A Whipple, Keyata Thompson, Amanda E Boggs, Jana Slovic, Edward H Cho, Michael A Matrone, Stuart S Martin
    Abstract:

    During metastasis, invading cells produce various actin-based membrane protrusions that promote directional migration and proteolysis of extracellular matrix (ECM). Observations of actin staining within thin, tubulin-based microtentacle (McTN) protrusions in suspended MDA-MB-231 tumor cells, prompted an investigation of whether McTNs are structural or functional analogs of Invadopodia. We show here that MDA-MB-231 cells are capable of producing Invadopodia and McTNs, both of which contain F-actin. Invadopodium formation was enhanced by the expression of a constitutively active c-Src kinase, and repressed by the expression of dominant-negative, catalytically inactive form of c-Src. In contrast, expression of inactive c-Src significantly increased McTN formation. Direct inhibition of c-Src with the SU6656 inhibitor compound also significantly enhanced McTN formation, but suppressed Invadopodia, including the appearance of F-actin cores and phospho-cortactin foci, as well as completely blocking focal degradation of ECM. In addition, silencing of Tks5 in Src-transformed fibroblasts blocked Invadopodia without affecting McTNs. Genetic modification of c-Src activity that promoted McTN formation augmented capillary retention of circulating tumor cells in vivo and rapid re-attachment of suspended cells in vitro, even though Invadopodia were strongly suppressed. These results indicate that McTNs are capable of enhancing tumor cell reattachment, even in the absence of Tks5 and active Src, and define separate cytoskeletal mechanisms and functions for McTNs and Invadopodia.

  • tumor suppressor function of syk in human mcf10a in vitro and normal mouse mammary epithelium in vivo
    PLOS ONE, 2009
    Co-Authors: You Me Sung, Francoise Seilliermoiseiwitsch, Junfeng Sun, Duane Mueller, Kinza Sentissi, Peter D Johnson, Elana Urbach, Michael D Johnson, Susette C Mueller
    Abstract:

    The normal function of Syk in epithelium of the developing or adult breast is not known, however, Syk suppresses tumor growth, invasion, and metastasis in breast cancer cells. Here, we demonstrate that in the mouse mammary gland, loss of one Syk allele profoundly increases proliferation and ductal branching and invasion of epithelial cells through the mammary fat pad during puberty. Mammary carcinomas develop by one year. Syk also suppresses proliferation and invasion in vitro. siRNA or shRNA knockdown of Syk in MCF10A breast epithelial cells dramatically increased proliferation, anchorage independent growth, cellular motility, and invasion, with formation of functional, extracellular matrix-degrading Invadopodia. Morphological and gene microarray analysis following Syk knockdown revealed a loss of luminal and differentiated epithelial features with epithelial to mesenchymal transition and a gain in Invadopodial cell surface markers CD44, CD49F, and MMP14. These results support the role of Syk in limiting proliferation and invasion of epithelial cells during normal morphogenesis, and emphasize the critical role of Syk as a tumor suppressor for breast cancer. The question of breast cancer risk following systemic anti-Syk therapy is raised since only partial loss of Syk was sufficient to induce mammary carcinomas.

  • the role of the exocyst in matrix metalloproteinase secretion and actin dynamics during tumor cell Invadopodia formation
    Molecular Biology of the Cell, 2009
    Co-Authors: Jianglan Liu, Vira V. Artym, Susette C Mueller, Peng Yue, Wei Guo
    Abstract:

    Invadopodia are actin-rich membrane protrusions formed by tumor cells that degrade the extracellular matrix for invasion. Invadopodia formation involves membrane protrusions driven by Arp2/3-mediat...

John S Condeelis - One of the best experts on this subject based on the ideXlab platform.

  • phosphorylated cortactin recruits vav2 guanine nucleotide exchange factor to activate rac3 and promote Invadopodial function in invasive breast cancer cells
    Molecular Biology of the Cell, 2017
    Co-Authors: Brian Rosenberg, John S Condeelis, Christopher C Mader, Hava Gilhenn, Tiffany L Halo, Taofei Yin, Kazuya Machida, Anthony J Koleske
    Abstract:

    Breast carcinoma cells use specialized, actin-rich protrusions called Invadopodia to degrade and invade through the extracellular matrix. Phosphorylation of the actin nucleation-promoting factor and actin-stabilizing protein cortactin downstream of the epidermal growth factor receptor-Src-Arg kinase cascade is known to be a critical trigger for invadopodium maturation and subsequent cell invasion in breast cancer cells. The functions of cortactin phosphorylation in this process, however, are not completely understood. We identify the Rho-family guanine nucleotide exchange factor Vav2 in a comprehensive screen for human SH2 domains that bind selectively to phosphorylated cortactin. We demonstrate that the Vav2 SH2 domain binds selectively to phosphotyrosine-containing peptides corresponding to cortactin tyrosines Y421 and Y466 but not to Y482. Mutation of the Vav2 SH2 domain disrupts its recruitment to Invadopodia, and an SH2-domain mutant form of Vav2 cannot support efficient matrix degradation in invasive MDA-MB-231 breast cancer cells. We show that Vav2 function is required for promoting invadopodium maturation and consequent actin polymerization, matrix degradation, and invasive migratory behavior. Using biochemical assays and a novel Rac3 biosensor, we show that Vav2 promotes Rac3 activation at Invadopodia. Rac3 knockdown reduces matrix degradation by Invadopodia, whereas a constitutively active Rac3 can rescue the deficits in invadopodium function in Vav2-knockdown cells. Together these data indicate that phosphorylated cortactin recruits Vav2 to activate Rac3 and promote Invadopodial maturation in invasive breast cancer cells.

  • abstract b05 invadopodium formation is enriched in perivascular niches and leads to cancer cell intravasation
    Cancer Research, 2016
    Co-Authors: Aviv Bergman, John S Condeelis, Bojana Gligorijevic
    Abstract:

    Invadopodia are actin-rich protrusions used for cancer cell movement across obstacles. In the past, they were mostly studied in cell cultures, where they form in contact with extracellular matrix. Our past work has suggested that elimination of key molecular components of Invadopodia in cancer cells can lead to elimination of metastasis. In the current study, we use high resolution multiphoton microscopy to understand how the tumor microenvironment controls formation of Invadopodia, followed by cancer cell locomotion, intravasation and metastasis. We demonstrate that the motile cancer cells in breast carcinoma in vivo can exhibit either invadopodium-related, slow locomotion or invadopodium-independent rapid locomotion along collagen fibers. These two phenotypes occur in spatially separate tumor regions, the location of which can be predicted from measurements of microenvironmental features. For example, cancer cells form Invadopodia predominantly in perivascular niches, regions surrounding major blood vessels and characterized by cross-linked collagen fibers and dense macrophages. Invadopodia in vivo extend towards blood vessels and their density decreases with blood vessel distance. We observe that only cells with Invadopodia are capable of intravasation, while fast locomotion results in cell dispersal throughout tumor. Further, either invadopodium-specific TkS5 knock-down or short-term inhibition of metalloproteases eliminate Invadopodia and stop dissemination to lung. Interestingly, external modulation of collagen cross-linking also inhibits Invadopodia, switching the cancer cell phenotype to fast locomotion. Our results suggest that the perivascular niche is the site of invadopodium formation and cancer cell intravasation. Moreover, Invadopodia and hence metastasis, can be inhibited by external modulation of the microenvironment. In summary, the presence of Invadopodia is a great candidate for a novel early-metastasis marker, while their elimination via microenvironment modulation has therapeutic potential. Citation Format: Aviv Bergman, John Condeelis, Bojana Gligorijevic. Invadopodium formation is enriched in perivascular niches and leads to cancer cell intravasation. [abstract]. In: Proceedings of the Fourth AACR International Conference on Frontiers in Basic Cancer Research; 2015 Oct 23-26; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2016;76(3 Suppl):Abstract nr B05.

  • microrna 375 suppresses extracellular matrix degradation and Invadopodial activity in head and neck squamous cell carcinoma
    Archives of Pathology & Laboratory Medicine, 2015
    Co-Authors: Lizandra Jimenez, Ved P Sharma, Geoffrey Childs, Michael B Prystowsky, Thomas J Ow, Thomas M. Harris, John S Condeelis, Jeffrey E Segall
    Abstract:

    CONTEXT: Head and neck squamous cell carcinoma (HNSCC) is a highly invasive cancer with an association with locoregional recurrence and lymph node metastasis. We have previously reported that low microRNA-375 (miR-375) expression levels correlate with poor patient survival, increased locoregional recurrence, and distant metastasis. Increasing miR-375 expression in HNSCC cell lines to levels found in normal cells results in suppressed invasive properties. HNSCC invasion is mediated in part by Invadopodia-associated degradation of the extracellular matrix. OBJECTIVE: To determine whether elevated miR-375 expression in HNSCC cell lines also affects Invadopodia formation and activity. DESIGN: For evaluation of the matrix degradation properties of the HNSCC lines, an Invadopodial matrix degradation assay was used. The total protein levels of Invadopodia-associated proteins were measured by Western blot analyses. Immunoprecipitation experiments were conducted to evaluate the tyrosine phosphorylation state of cortactin. Human protease arrays were used for the detection of the secreted proteases. Quantitative real time-polymerase chain reaction measurements were used to evaluate the messenger RNA (mRNA) expression of the commonly regulated proteases. RESULTS: Increased miR-375 expression in HNSCC cells suppresses extracellular matrix degradation and reduces the number of mature Invadopodia. Higher miR-375 expression does not reduce cellular levels of selected Invadopodia-associated proteins, nor is tyrosine phosphorylation of cortactin altered. However, HNSCC cells with higher miR-375 expression had significant reductions in the mRNA expression levels and secreted levels of specific proteases. CONCLUSIONS: MicroRNA-375 regulates Invadopodia maturation and function potentially by suppressing the expression and secretion of proteases.

  • abstract 185 microrna 375 suppresses extracellular matrix degradation and Invadopodial activity in head and neck squamous cell carcinoma
    Cancer Research, 2015
    Co-Authors: Lizandra Jimenez, Ved P Sharma, Geoffrey Childs, Michael B Prystowsky, Thomas J Ow, Thomas M. Harris, John S Condeelis, Jeffrey E Segall
    Abstract:

    Context.— Head and neck squamous cell carcinoma (HNSCC) is a highly invasive cancer with an association with locoregional recurrence and lymph node metastasis. We have previously reported that low microRNA-375 (miR-375) expression levels correlate with poor patient survival, increased locoregional recurrence, and distant metastasis. Increasing miR-375 expression in HNSCC cell lines to levels found in normal cells results in suppressed invasive properties. HNSCC invasion is mediated in part by Invadopodia-associated degradation of the extracellular matrix. Objective.— To determine whether elevated miR-375 expression in HNSCC cell lines also affects Invadopodia formation and activity. Design.— For evaluation of the matrix degradation properties of the HNSCC lines, an Invadopodial matrix degradation assay was used. The total protein levels of Invadopodia-associated proteins were measured by Western blot analyses. Immunoprecipitation experiments were conducted to evaluate the tyrosine phosphorylation state of...

  • talin regulates moesin nhe 1 recruitment to Invadopodia and promotes mammary tumor metastasis
    Journal of Cell Biology, 2014
    Co-Authors: Brian T Beaty, Ved P Sharma, Jose Javier Bravocordero, Yarong Wang, Louis Hodgson, Veronika Miskolci, John S Condeelis
    Abstract:

    Invadopodia are actin-rich protrusions that degrade the extracellular matrix and are required for stromal invasion, intravasation, and metastasis. The role of the focal adhesion protein talin in regulating these structures is not known. Here, we demonstrate that talin is required for Invadopodial matrix degradation and three-dimensional extracellular matrix invasion in metastatic breast cancer cells. The sodium/hydrogen exchanger 1 (NHE-1) is linked to the cytoskeleton by ezrin/radixin/moesin family proteins and is known to regulate invadopodium-mediated matrix degradation. We show that the talin C terminus binds directly to the moesin band 4.1 ERM (FERM) domain to recruit a moesin–NHE-1 complex to Invadopodia. Silencing talin resulted in a decrease in cytosolic pH at Invadopodia and blocked cofilin-dependent actin polymerization, leading to impaired invadopodium stability and matrix degradation. Furthermore, talin is required for mammary tumor cell motility, intravasation, and spontaneous lung metastasis in vivo. Thus, our findings provide a novel understanding of how intracellular pH is regulated and a molecular mechanism by which talin enhances tumor cell invasion and metastasis.

Jeffrey E Segall - One of the best experts on this subject based on the ideXlab platform.

  • tc10 regulates breast cancer invasion and metastasis by controlling membrane type 1 matrix metalloproteinase at Invadopodia
    Unknown Journal, 2020
    Co-Authors: Maren Hulsemann, Jeffrey E Segall, Sara K Donnelly, Polina V Verkhusha, Serena P H Mao, Louis Hodgson
    Abstract:

    Abstract During breast cancer metastasis, cancer cell invasion is driven by actin-rich protrusions called Invadopodia, which mediate the extracellular matrix degradation required for the success of the invasive cascade. In this study, we demonstrated that TC10, a member of a Cdc42 subfamily of p21 small GTPases, regulates the membrane type 1 matrix metalloproteinase (MT1-MMP)-driven extracellular matrix degradation at Invadopodia. We show that TC10 is required for the plasma membrane surface exposure of MT1-MMP at Invadopodia. By utilizing our new Forster resonance energy transfer (FRET) biosensor, we demonstrated the p190RhoGAP-dependent regulation of spatiotemporal TC10 activity at Invadopodia. We identified a pathway that regulates TC10 activity and function at Invadopodia through the activation of p190RhoGAP and the downstream interacting effector Exo70 at the Invadopodia sites. Our findings reveal the role of a previously unknown regulator of vesicular fusion at Invadopodia, TC10, on the invasive potential of breast cancer cells during invasion and metastasis.

  • microrna 375 suppresses extracellular matrix degradation and Invadopodial activity in head and neck squamous cell carcinoma
    Archives of Pathology & Laboratory Medicine, 2015
    Co-Authors: Lizandra Jimenez, Ved P Sharma, Geoffrey Childs, Michael B Prystowsky, Thomas J Ow, Thomas M. Harris, John S Condeelis, Jeffrey E Segall
    Abstract:

    CONTEXT: Head and neck squamous cell carcinoma (HNSCC) is a highly invasive cancer with an association with locoregional recurrence and lymph node metastasis. We have previously reported that low microRNA-375 (miR-375) expression levels correlate with poor patient survival, increased locoregional recurrence, and distant metastasis. Increasing miR-375 expression in HNSCC cell lines to levels found in normal cells results in suppressed invasive properties. HNSCC invasion is mediated in part by Invadopodia-associated degradation of the extracellular matrix. OBJECTIVE: To determine whether elevated miR-375 expression in HNSCC cell lines also affects Invadopodia formation and activity. DESIGN: For evaluation of the matrix degradation properties of the HNSCC lines, an Invadopodial matrix degradation assay was used. The total protein levels of Invadopodia-associated proteins were measured by Western blot analyses. Immunoprecipitation experiments were conducted to evaluate the tyrosine phosphorylation state of cortactin. Human protease arrays were used for the detection of the secreted proteases. Quantitative real time-polymerase chain reaction measurements were used to evaluate the messenger RNA (mRNA) expression of the commonly regulated proteases. RESULTS: Increased miR-375 expression in HNSCC cells suppresses extracellular matrix degradation and reduces the number of mature Invadopodia. Higher miR-375 expression does not reduce cellular levels of selected Invadopodia-associated proteins, nor is tyrosine phosphorylation of cortactin altered. However, HNSCC cells with higher miR-375 expression had significant reductions in the mRNA expression levels and secreted levels of specific proteases. CONCLUSIONS: MicroRNA-375 regulates Invadopodia maturation and function potentially by suppressing the expression and secretion of proteases.

  • abstract 185 microrna 375 suppresses extracellular matrix degradation and Invadopodial activity in head and neck squamous cell carcinoma
    Cancer Research, 2015
    Co-Authors: Lizandra Jimenez, Ved P Sharma, Geoffrey Childs, Michael B Prystowsky, Thomas J Ow, Thomas M. Harris, John S Condeelis, Jeffrey E Segall
    Abstract:

    Context.— Head and neck squamous cell carcinoma (HNSCC) is a highly invasive cancer with an association with locoregional recurrence and lymph node metastasis. We have previously reported that low microRNA-375 (miR-375) expression levels correlate with poor patient survival, increased locoregional recurrence, and distant metastasis. Increasing miR-375 expression in HNSCC cell lines to levels found in normal cells results in suppressed invasive properties. HNSCC invasion is mediated in part by Invadopodia-associated degradation of the extracellular matrix. Objective.— To determine whether elevated miR-375 expression in HNSCC cell lines also affects Invadopodia formation and activity. Design.— For evaluation of the matrix degradation properties of the HNSCC lines, an Invadopodial matrix degradation assay was used. The total protein levels of Invadopodia-associated proteins were measured by Western blot analyses. Immunoprecipitation experiments were conducted to evaluate the tyrosine phosphorylation state of...

  • molecular mechanisms of invadopodium formation the role of the n wasp arp2 3 complex pathway and cofilin
    Journal of Cell Biology, 2005
    Co-Authors: Hideki Yamaguchi, Jeffrey E Segall, Marc Symons, Robert J Eddy, Mike Lorenz, Stephan J Kempiak, Corina Sarmiento, Salvatore J Coniglio, Hiroaki Miki
    Abstract:

    Invadopodia are actin-rich membrane protrusions with a matrix degradation activity formed by invasive cancer cells. We have studied the molecular mechanisms of invadopodium formation in metastatic carcinoma cells. Epidermal growth factor (EGF) receptor kinase inhibitors blocked invadopodium formation in the presence of serum, and EGF stimulation of serum-starved cells induced invadopodium formation. RNA interference and dominant-negative mutant expression analyses revealed that neural WASP (N-WASP), Arp2/3 complex, and their upstream regulators, Nck1, Cdc42, and WIP, are necessary for invadopodium formation. Time-lapse analysis revealed that Invadopodia are formed de novo at the cell periphery and their lifetime varies from minutes to several hours. Invadopodia with short lifetimes are motile, whereas long-lived Invadopodia tend to be stationary. Interestingly, suppression of cofilin expression by RNA interference inhibited the formation of long-lived Invadopodia, resulting in formation of only short-lived Invadopodia with less matrix degradation activity. These results indicate that EGF receptor signaling regulates invadopodium formation through the N-WASP–Arp2/3 pathway and cofilin is necessary for the stabilization and maturation of Invadopodia.

Kiyoko Fukami - One of the best experts on this subject based on the ideXlab platform.

  • phosphoinositide 3 kinase signaling pathway mediated by p110α regulates Invadopodia formation
    Journal of Cell Biology, 2011
    Co-Authors: Hideki Yamaguchi, Shuhei Yoshida, Emi Muroi, Nachi Yoshida, Masahiro Kawamura, Zen Kouchi, Yoshikazu Nakamura, Ryuichi Sakai, Kiyoko Fukami
    Abstract:

    Invadopodia are extracellular matrix–degrading protrusions formed by invasive cancer cells that are thought to function in cancer invasion. Although many Invadopodia components have been identified, signaling pathways that link extracellular stimuli to Invadopodia formation remain largely unknown. We investigate the role of phosphoinositide 3-kinase (PI3K) signaling during Invadopodia formation. We find that in human breast cancer cells, both Invadopodia formation and degradation of a gelatin matrix were blocked by treatment with PI3K inhibitors or sequestration of D-3 phosphoinositides. Functional analyses revealed that among the PI3K family proteins, the class I PI3K catalytic subunit p110α, a frequently mutated gene product in human cancers, was selectively involved in Invadopodia formation. The expression of p110α with cancerous mutations promoted Invadopodia-mediated invasive activity. Furthermore, knockdown or inhibition of PDK1 and Akt, downstream effectors of PI3K signaling, suppressed Invadopodia formation induced by p110α mutants. These data suggest that PI3K signaling via p110α regulates Invadopodia-mediated invasion of breast cancer cells.

  • lipid rafts and caveolin 1 are required for Invadopodia formation and extracellular matrix degradation by human breast cancer cells
    Cancer Research, 2009
    Co-Authors: Hideki Yamaguchi, Shuhei Yoshida, Zen Kouchi, Yoshikazu Nakamura, Yukiko Takeo, Kiyoko Fukami
    Abstract:

    Invadopodia are ventral membrane protrusions through which invasive cancer cells degrade the extracellular matrix. They are thought to function in the migration of cancer cells through tissue barriers, which is necessary for cancer invasion and metastasis. Although many protein components of Invadopodia have been identified, the organization and the role of membrane lipids in Invadopodia are not well understood. In this study, the role of lipid rafts, which are cholesterol-enriched membrane microdomains, in the assembly and function of Invadopodia in human breast cancer cells was investigated. Lipid rafts are enriched, internalized, and dynamically trafficked at Invadopodia sites. Perturbation of lipid raft formation due to depleting or sequestering membrane cholesterol blocked the Invadopodia-mediated degradation of the gelatin matrix. Caveolin-1 (Cav-1), a resident protein of lipid rafts and caveolae, accumulates at Invadopodia and colocalizes with the internalized lipid raft membranes. Membrane type 1 matrix metalloproteinase (MT1-MMP), a matrix proteinase associated with Invadopodia, is localized at lipid raft-enriched membrane fractions and cotrafficked and colocalized with Cav-1 at Invadopodia. The small interfering RNA–mediated silencing of Cav-1 inhibited the Invadopodia-mediated and MT1-MMP–dependent degradation of the gelatin matrix. Furthermore, Cav-1 and MT1-MMP are coexpressed in invasive human breast cancer cell lines that have an ability to form Invadopodia. These results indicate that Invadopodia are the sites where enrichment and trafficking of lipid rafts occur and that Cav-1 is an essential regulator of MT1-MMP function and Invadopodia-mediated breast cancer cell invasion. [Cancer Res 2009;69(22):8594–602]

  • lipid rafts and caveolin 1 are required for Invadopodia formation and extracellular matrix degradation by human breast cancer cells
    Cancer Research, 2009
    Co-Authors: Hideki Yamaguchi, Shuhei Yoshida, Zen Kouchi, Yoshikazu Nakamura, Yukiko Takeo, Kiyoko Fukami
    Abstract:

    Invadopodia are ventral membrane protrusions through which invasive cancer cells degrade the extracellular matrix. They are thought to function in the migration of cancer cells through tissue barriers, which is necessary for cancer invasion and metastasis. Although many protein components of Invadopodia have been identified, the organization and the role of membrane lipids in Invadopodia are not well understood. In this study, the role of lipid rafts, which are cholesterol-enriched membrane microdomains, in the assembly and function of Invadopodia in human breast cancer cells was investigated. Lipid rafts are enriched, internalized, and dynamically trafficked at Invadopodia sites. Perturbation of lipid raft formation due to depleting or sequestering membrane cholesterol blocked the Invadopodia-mediated degradation of the gelatin matrix. Caveolin-1 (Cav-1), a resident protein of lipid rafts and caveolae, accumulates at Invadopodia and colocalizes with the internalized lipid raft membranes. Membrane type 1 matrix metalloproteinase (MT1-MMP), a matrix proteinase associated with Invadopodia, is localized at lipid raft-enriched membrane fractions and cotrafficked and colocalized with Cav-1 at Invadopodia. The small interfering RNA-mediated silencing of Cav-1 inhibited the Invadopodia-mediated and MT1-MMP-dependent degradation of the gelatin matrix. Furthermore, Cav-1 and MT1-MMP are coexpressed in invasive human breast cancer cell lines that have an ability to form Invadopodia. These results indicate that Invadopodia are the sites where enrichment and trafficking of lipid rafts occur and that Cav-1 is an essential regulator of MT1-MMP function and Invadopodia-mediated breast cancer cell invasion.