Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 6, No. 5 ( 2016-05-01), p. 516-531
    Abstract: Fibronectin (FN) is a major component of the tumor microenvironment, but its role in promoting metastasis is incompletely understood. Here, we show that FN gradients elicit directional movement of breast cancer cells, in vitro and in vivo. Haptotaxis on FN gradients requires direct interaction between α5β1 integrin and MENA, an actin regulator, and involves increases in focal complex signaling and tumor cell–mediated extracellular matrix (ECM) remodeling. Compared with MENA, higher levels of the prometastatic MENAINV isoform associate with α5, which enables 3-D haptotaxis of tumor cells toward the high FN concentrations typically present in perivascular space and in the periphery of breast tumor tissue. MENAINV and FN levels were correlated in two breast cancer cohorts, and high levels of MENAINV were significantly associated with increased tumor recurrence as well as decreased patient survival. Our results identify a novel tumor cell–intrinsic mechanism that promotes metastasis through ECM remodeling and ECM-guided directional migration. Significance: Here, we provide new insight into how tumor cell:ECM interactions generate signals and structures that promote directed tumor cell migration, a critical component of metastasis. Our results identify a tumor cell–intrinsic mechanism driven by the actin regulatory protein MENA that promotes ECM remodeling and haptotaxis along FN gradients. Cancer Discov; 6(5); 516–31. ©2016 AACR. See related commentary by Santiago-Medina and Yang, p. 474. This article is highlighted in the In This Issue feature, p. 461
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1988-1988
    Abstract: To metastasize and disseminate, carcinoma cells must invade the surrounding extracellular matrix (ECM) and migrate to reach blood or lymphatic vessels. Collagen (CN) and fibronectin (FN), two components of the ECM surrounding tumors, are highly expressed in metastatic tumors and their patterns of expression can correlate with metastatic outcome in patient samples. Mena, an actin regulatory protein, is upregulated in breast cancer and is alternatively spliced to produce protein isoforms with distinct functions during tumor progression. The invasion-specific MenaINV isoform increases metastasis by potentiating tumor cell motility and invasion responses to various growth factors. All Mena isoforms bind directly to the α5 subunit of the α5β1 integrin, an FN receptor. We hypothesized that the interaction between MenaINV and α5β1 plays an important role in integrating signals from the ECM as well as growth factors to drive invasion. MDA-MB-231 breast cancer cells expressing MenaINV showed increased adhesion to FN alone as well as mixture of FN and CN, compared to control cells. MenaINV cells also had an increased number of α5-positive adhesions as well as increased signalling at focal adhesions as measured by phospho-Paxilin, compared to control. In 3D invasion assays, addition of FN to a CN gel drove invasion of MenaINV-expressing cells even in the absence of any growth factor ligand, an effect driven by its interaction with α5 and dependent on signalling via EGFR and Met. Furthermore, addition of FN caused an even greater potentiation of EGF-induced invasion by MenaINV, an effect that was dependent upon its interaction with α5. Next, we examined how MenaINV affects tumor cell responses to a gradient of FN within a 3D collagen gel. Control cells failed to respond to an FN gradient, while cells expressing MenaINV migrated preferentially towards FN and significantly reorganized both the collagen and FN surrounding them. Deletion of the α5 binding site or treatment with an α5 function blocking antibody ablated the effects MenaINV on movement towards a FN gradient as well ECM reorganization. Overall, these results suggest that some aspects of the pro-metastatic effects of MenaINV involve its ability to bind α5 and regulate bidirectional signaling with FN and growth factors in the tumor microenvironment. Citation Format: Madeleine J. Oudin, Liliane C. Broye, Alisha Lussiez, Sreeja B. Asokan, Miles A. Miller, Douglas A. Lauffenburger, James E. Bear, Frank B. Gertler. MenaINV interaction with α5β1 promotes tumor cell invasion in response to gradients of growth factors and fibronectin. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1988. doi:10.1158/1538-7445.AM2014-1988
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 3_Supplement ( 2013-02-01), p. C41-C41
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 3_Supplement ( 2013-02-01), p. C41-C41
    Abstract: Mena, an actin regulatory protein, is upregulated in human breast cancer and its expression is associated with increased metastatic progression. An invasion-specific isoform, MenaINV, is upregulated selectively in invasive tumor cells and increases invasion and metastasis by potentiating tumor cell responses to EGF. MenaINV causes dysregulation of the tyrosine phosphatase PTP1B, leading to increased phosphorylation of EGFR and other downstream targets. We investigated whether MenaINV could similarly promote invasion through dysregulation of signaling downstream of other receptor tyrosine kinases (RTKs). The behavior of MDA-MB-231 triple negative breast cancer cells stably expressing MenaINV or the other cannonical, broadly expressed Mena lacking the “INV” exon was measured after growth factor treatment in two migration assays: a 2D protrusion assay on matrigel and collagen coated coverslips and a 3D invasion assay into collagen gels. We found that expression of MenaINV sensitized responses to other EGFR ligands including HB-EGF and amphiregulin. In addition, MenaINV expression enhanced responses mediated by two other RTKs associated with tumor progression, Met and IGFR, both known PTP1B substrates. Cells expressing MenaINV respond to 20-fold lower concentrations HGF and IGF than control cells. However, there was no effect of MenaINV on responses to the ERBB3 ligand neuregulin and the chemokine SDF-1. Recently we reported that Mena can regulate cell motility in fibroblasts via its interaction with α5β1 integrin, which is known to co-traffic with EGFR and other RTKs to regulate invasion in tumor cells expressing mutant p53 or upon inhibition of αvβ3. We asked whether the direct interaction of Mena with α5β1 also contributes to invasion. Disrupting the interaction between the Mena isoforms and α5β1 in MDA-MB-231 cells affected how cells responded to EGF in a 3D collagen gel. In addition, fibroblasts that were null for Mena were unable to respond to a fibronectin gradient in a haptotaxis assay. Overall, these results suggest that the effects of MenaINV on invasion and metastasis could be mediated via different RTKs, as well as through direct interaction with integrins and the extracellular matrix. Experiments are currently underway to investigate how MenaINV regulates signalling downstream of these RTKs and integrins, to understand the molecular mechanism underlying the pro-metastatic effect of MenaINV. Citation Format: Madeleine J. Oudin, Shannon K. Hughes-Alford, Miles Miller, Sreeja B. Asokan, James E. Bear, Doug A. Lauffenburger, Frank B. Gertler. MenaINV dysregulation of receptor tyrosine kinase signaling. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr C41.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Cell Biology, Rockefeller University Press, Vol. 203, No. 6 ( 2013-12-23), p. 907-916
    Abstract: Arp2/3-branched actin is critical for cytoskeletal dynamics and cell migration. However, perturbations and diseases affecting this network have phenotypes that cannot be fully explained by cell-autonomous effects. In this paper, we report nonautonomous effects of Arp2/3 depletion. We show that, upon Arp2/3 depletion, the expression of numerous genes encoding secreted factors, including chemokines, growth factors, and matrix metalloproteases, was increased, a signature resembling the senescence-associated secretory phenotype. These factors affected epidermal growth factor chemotaxis in a nonautonomous way, resolving the recent contradictions about the role of Arp2/3 in chemotaxis. We demonstrate that these genes were activated by nuclear factor κB via a CCM2–MEKK3 pathway that has been implicated in hyperosmotic stress signaling. Consistent with this, Arp2/3-depleted cells showed misregulation of volume control and reduced actin in the submembranous cortex. The defects in osmotic signaling in the Arp2/3-depleted cells can be rescued by hypoosmotic treatment. Thus, perturbations of Arp2/3 have nonautonomous effects that should be considered when evaluating experimental manipulations and diseases affecting the Arp2/3-actin cytoskeleton.
    Type of Medium: Online Resource
    ISSN: 1540-8140 , 0021-9525
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2013
    detail.hit.zdb_id: 1421310-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Cell Biology, Rockefeller University Press, Vol. 207, No. 2 ( 2014-10-27), p. 299-315
    Abstract: Somatic inactivation of the serine/threonine kinase gene STK11/LKB1/PAR-4 occurs in a variety of cancers, including ∼10% of melanoma. However, how the loss of LKB1 activity facilitates melanoma invasion and metastasis remains poorly understood. In LKB1-null cells derived from an autochthonous murine model of melanoma with activated Kras and Lkb1 loss and matched reconstituted controls, we have investigated the mechanism by which LKB1 loss increases melanoma invasive motility. Using a microfluidic gradient chamber system and time-lapse microscopy, in this paper, we uncover a new function for LKB1 as a directional migration sensor of gradients of extracellular matrix (haptotaxis) but not soluble growth factor cues (chemotaxis). Systematic perturbation of known LKB1 effectors demonstrated that this response does not require canonical adenosine monophosphate–activated protein kinase (AMPK) activity but instead requires the activity of the AMPK-related microtubule affinity-regulating kinase (MARK)/PAR-1 family kinases. Inhibition of the LKB1–MARK pathway facilitated invasive motility, suggesting that loss of the ability to sense inhibitory matrix cues may promote melanoma invasion.
    Type of Medium: Online Resource
    ISSN: 1540-8140 , 0021-9525
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2014
    detail.hit.zdb_id: 1421310-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 3_Supplement ( 2013-02-01), p. C14-C14
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 3_Supplement ( 2013-02-01), p. C14-C14
    Abstract: Germline mutations in the serine/threonine kinase STK11/LKB1 are associated with Peutz-Jehgers Syndrome, which is characterized by hyperpigmentation of the oral mucosa. Inactivating somatic mutations occur in approximately 10-20% of melanomas; however, how the loss of LKB1 facilitates melanoma invasion remains poorly understood. Using cell lines derived from simultaneous activation of KRas and inactivation of LKB1 in melanocytes, we have investigated melanoma migration upon reconstitution with LKB1. Reexpression of LKB1 diminishes migration during wound healing, spheroid outgrowth into 3D collagen, and overall single cell speed in random motility assays. Furthermore, the formation of invadopodia is independent of LKB1 status in both human and mouse melanomas. Interestingly, using microfluidic devices we have found that loss of LKB1 abrogates the ability of cells to respond to gradients of extracellular matrix (haptotaxis) but does not impair their ability to chemotax to EGF. We have also recently developed a model of orthotopic implantation of multicellular tumor spheroids into the dermis of the mouse ear skin and have validated this approach by recapitulating the finding that LKB1 limits tumorigenesis. We are using this model to image local invasion in vivo by multiphoton microscopy and are currently examining the intriguing hypothesis that loss of extracellular matrix sensing is one aspect that contributes to metastatic migration. Citation Format: Keefe T. Chan, Sreeja B. Asokan, Tao Bo, Matthew E. Berginski, Wenjin Liu, Shelly D. Cochran, Norman E. Sharpless, James E. Bear. Loss of haptotaxis facilitates invasion in LKB1-deficient melanoma. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Invasion and Metastasis; Jan 20-23, 2013; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2013;73(3 Suppl):Abstract nr C14.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Rockefeller University Press ; 2017
    In:  Journal of Cell Biology Vol. 216, No. 9 ( 2017-09-04), p. 2877-2889
    In: Journal of Cell Biology, Rockefeller University Press, Vol. 216, No. 9 ( 2017-09-04), p. 2877-2889
    Abstract: Nonmuscle myosin II (NMII) is uniquely responsible for cell contractility and thus defines multiple aspects of cell behavior. To generate contraction, NMII molecules polymerize into bipolar minifilaments. Different NMII paralogs are often coexpressed in cells and can copolymerize, suggesting that they may cooperate to facilitate cell motility. However, whether such cooperation exists and how it may work remain unknown. We show that copolymerization of NMIIA and NMIIB followed by their differential turnover leads to self-sorting of NMIIA and NMIIB along the front–rear axis, thus producing a polarized actin–NMII cytoskeleton. Stress fibers newly formed near the leading edge are enriched in NMIIA, but over time, they become progressively enriched with NMIIB because of faster NMIIA turnover. In combination with retrograde flow, this process results in posterior accumulation of more stable NMIIB-rich stress fibers, thus strengthening cell polarity. By copolymerizing with NMIIB, NMIIA accelerates the intrinsically slow NMIIB dynamics, thus increasing cell motility and traction and enabling chemotaxis.
    Type of Medium: Online Resource
    ISSN: 0021-9525 , 1540-8140
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2017
    detail.hit.zdb_id: 1421310-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2019
    In:  Integrative Biology Vol. 11, No. 6 ( 2019-06-01), p. 280-292
    In: Integrative Biology, Oxford University Press (OUP), Vol. 11, No. 6 ( 2019-06-01), p. 280-292
    Abstract: We used particle-based computer simulations to study the emergent properties of the actomyosin cytoskeleton. Our model accounted for biophysical interactions between filamentous actin and non-muscle myosin II and was motivated by recent experiments demonstrating that spatial regulation of myosin activity is required for fibroblasts responding to spatial gradients of platelet derived growth factor (PDGF) to undergo chemotaxis. Our simulations revealed the spontaneous formation of actin asters, consistent with the punctate actin structures observed in chemotacting fibroblasts. We performed a systematic analysis of model parameters to identify biochemical steps in myosin activity that significantly affect aster formation and performed simulations in which model parameter values vary spatially to investigate how the model responds to chemical gradients. Interestingly, spatial variations in motor stiffness generated time-dependent behavior of the actomyosin network, in which actin asters continued to spontaneously form and dissociate in different regions of the gradient. Our results should serve as a guide for future experimental investigations.
    Type of Medium: Online Resource
    ISSN: 1757-9708
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2019
    detail.hit.zdb_id: 2480063-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Rockefeller University Press ; 2015
    In:  Journal of Cell Biology Vol. 209, No. 6 ( 2015-06-22), p. 803-812
    In: Journal of Cell Biology, Rockefeller University Press, Vol. 209, No. 6 ( 2015-06-22), p. 803-812
    Abstract: The lamellipodium is an important structure for cell migration containing branched actin nucleated via the Arp2/3 complex. The formation of branched actin is relatively well studied, but less is known about its disassembly and how this influences migration. GMF is implicated in both Arp2/3 debranching and inhibition of Arp2/3 activation. Modulation of GMFβ, a ubiquitous GMF isoform, by depletion or overexpression resulted in changes in lamellipodial dynamics, branched actin content, and migration. Acute pharmacological inhibition of Arp2/3 by CK-666, coupled to quantitative live-cell imaging of the complex, showed that depletion of GMFβ decreased the rate of branched actin disassembly. These data, along with mutagenesis studies, suggest that debranching (not inhibition of Arp2/3 activation) is a primary activity of GMFβ in vivo. Furthermore, depletion or overexpression of GMFβ disrupted the ability of cells to directionally migrate to a gradient of fibronectin (haptotaxis). These data suggest that debranching by GMFβ plays an important role in branched actin regulation, lamellipodial dynamics, and directional migration.
    Type of Medium: Online Resource
    ISSN: 0021-9525 , 1540-8140
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2015
    detail.hit.zdb_id: 1421310-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2016
    In:  Integrative Biology Vol. 8, No. 8 ( 2016-08-08), p. 894-903
    In: Integrative Biology, Oxford University Press (OUP), Vol. 8, No. 8 ( 2016-08-08), p. 894-903
    Type of Medium: Online Resource
    ISSN: 1757-9708
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2016
    detail.hit.zdb_id: 2480063-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages