GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 3148-3148
    Abstract: The goal of this study was to investigate a novel mechanism by which neural extracellular matrix (ECM) proteins regulate the invasive behavior of malignant gliomas, which are the most common primary brain tumors in adults. Invasion of glioma cells is facilitated by secreted metalloproteases that cleave ECM proteoglycans that inhibit cell migration. Surprisingly, glioma cells also produce these proteoglycans at higher levels than normal neural cells, which appears paradoxical. One of these proteoglycans, brevican (BCAN), is uniquely expressed in the CNS, is the most abundant chondroitin-sulfate proteoglycan in the adult brain ECM, and is upregulated in all low-grade gliomas and glioblastomas independently of their molecular subtype. The functions of brevican, both in normal neural cells or glioma cells, remain unknown and are assumed to be purely structural. We analyzed the signaling mechanisms of brevican using gain- and loss-of-function approaches in differentiated glioblastoma cell lines as well as tumor stem cells, combined with experiments of cell adhesion and invasion in vitro and in vivo. We discovered that a fragment of brevican, but not the full-length protein, interacts with cell-surface sulfatides and activates Src kinase, resulting in trans-activation of EGFR/MAPK signaling even in absence of the native EGFR ligands (TGF-alpha or EGF). Brevican-enhanced EGFR/MAPK activation resulted in increased cell adhesion -via fibronectin production- and motility, which were reversed using Src inhibitors or by treating the cells with aryl-sulfatase that removes sulfatides from the cell surface. Importantly, we observed that brevican secreted by glioblastoma cells was cleaved not only by these cells but also by normal astrocytes that were co-opted by the tumor cells. Absence of this cooperative effect was observed when tumors were implanted in an EGFR-deficient mouse model in which astrocytes did not process glioma-derived brevican, resulting in significantly reduced tumor dispersion. These results resolve the paradoxical production of "anti-migratory" proteoglycans by tumor cells; establish for the first time the entire signaling axis for the proteoglycan brevican in glioma cells; and reveal how this proteoglycan mediates a cooperative interaction between tumor cells and astrocytes that is needed for glioma invasion. These fundamental studies may be leveraged to advance novel anti-invasive strategies that could potentiate the efficacy of current glioma therapies. Citation Format: Somanath Kundu, Hosung Sim, Bin Hu, Mohan S. Nandhu, Russell T. Matthews, Mariano S. Viapiano. A novel mechanism of glioblastoma cell invasion regulated by a neural proteoglycan that activates Src/EGFR signaling and mediates tumor-astroglia cooperation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 3148.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 15 ( 2012-08-01), p. 3873-3885
    Abstract: Malignant gliomas are highly invasive and chemoresistant brain tumors with extremely poor prognosis. Targeting of the soluble factors that trigger invasion and resistance, therefore, could have a significant impact against the infiltrative glioma cells that are a major source of recurrence. Fibulin-3 is a matrix protein that is absent in normal brain but upregulated in gliomas and promotes tumor invasion by unknown mechanisms. Here, we show that fibulin-3 is a novel soluble activator of Notch signaling that antagonizes DLL3, an autocrine inhibitor or Notch, and promotes tumor cell survival and invasion in a Notch-dependent manner. Using a strategy for inducible knockdown, we found that controlled downregulation of fibulin-3 reduced Notch signaling and led to increased apoptosis, reduced self-renewal of glioblastoma-initiating cells, and impaired growth and dispersion of intracranial tumors. In addition, fibulin-3 expression correlated with expression levels of Notch-dependent genes and was a marker of Notch activation in patient-derived glioma samples. These findings underscore a major role for the tumor extracellular matrix in regulating glioma invasion and resistance to apoptosis via activation of the key Notch pathway. More importantly, this work describes a noncanonical, soluble activator of Notch in a cancer model and shows how Notch signaling can be reduced by targeting tumor-specific accessible molecules in the tumor microenvironment. Cancer Res; 72(15); 3873–85. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2353-2353
    Abstract: Malignant gliomas are the most common primary brain tumors and have extremely poor prognosis. Glioma cell infiltrate into normal neural tissue, escaping conventional chemo-radiotherapy and making tumor relapse almost unavoidable. We have recently characterized a novel extracellular matrix (ECM) protein named fibulin-3, which is highly expressed in gliomas but is absent in normal brain and rarely expressed in other solid tumors. We demonstrated that fibulin-3 regulates the expression of metalloproteases and promotes glioma cell invasion. In addition, this ECM protein increases the resistance of glioma cells to pro-apoptotic treatments, including starvation, oxidative stress and treatment with temozolomide. To further understand the mechanisms of fibulin-3 in gliomas, we investigated several candidate pathways but found no activation of EGFR, MAPK or Akt signaling. Interestingly, fibulin-3 bears significant homology with the Notch ligands from the Delta-like family, which prompted us to study the possible activation of this pathway, which is a critical anti-apoptotic pathway in cancer cells. Surprisingly, we found that fibulin-3 increased cleavage of endogenous Notch-1 in glioma cells as well as activation of a Notch-dependent reporter. In agreement, fibulin-3 expression regulated the levels of the Notch effector Hes-1 both in vitro and in vivo. Moreover, the pro-invasive effect of fibulin-3 in glioma cells was inhibited by pharmacological inhibition of Notch signaling or knockdown of Notch-1. This is potentially a novel mechanism by which tumor-associated ECM molecules can activate Notch signaling and modulate this key pro-tumoral pathway in gliomas. These results underscore the potential of fibulin-3 as a glioma-specific target in the tumor microenvironment. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2353. doi:10.1158/1538-7445.AM2011-2353
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Elsevier BV ; 2009
    In:  Journal of Biological Chemistry Vol. 284, No. 39 ( 2009-09), p. 26547-26556
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 284, No. 39 ( 2009-09), p. 26547-26556
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2009
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Molecular Cancer Research Vol. 7, No. 11 ( 2009-11-01), p. 1756-1770
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 7, No. 11 ( 2009-11-01), p. 1756-1770
    Abstract: Malignant gliomas are highly invasive tumors with an almost invariably rapid and lethal outcome. Surgery and chemoradiotherapy fail to remove resistant tumor cells that disperse within normal tissue, which are a major cause for disease progression and therapy failure. Infiltration of the neural parenchyma is a distinctive property of malignant gliomas compared with other solid tumors. Thus, glioma cells are thought to produce unique molecular changes that remodel the neural extracellular matrix and form a microenvironment permissive for their motility. Here, we describe the unique expression and proinvasive role of fibulin-3, a mesenchymal matrix protein specifically upregulated in gliomas. Fibulin-3 is downregulated in peripheral tumors and is thought to inhibit tumor growth. However, we found fibulin-3 highly upregulated in gliomas and cultured glioma cells, although the protein was undetectable in normal brain or cultured astrocytes. Overexpression and knockdown experiments revealed that fibulin-3 did not seem to affect glioma cell morphology or proliferation, but enhanced substrate-specific cell adhesion and promoted cell motility and dispersion in organotypic cultures. Moreover, orthotopic implantation of fibulin-3–overexpressing glioma cells resulted in diffuse tumors with increased volume and rostrocaudal extension compared with controls. Tumors and cultured cells overexpressing fibulin-3 also showed elevated expression and activity of matrix metalloproteases, such as MMP-2/MMP-9 and ADAMTS-5. Taken together, our results suggest that fibulin-3 has a unique expression and protumoral role in gliomas, and could be a potential target against tumor progression. Strategies against this glioma-specific matrix component could disrupt invasive mechanisms and restrict the dissemination of these tumors. (Mol Cancer Res 2009;7(11):1756–70)
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...