Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2015
    In:  Inflammatory Bowel Diseases Vol. 21, No. 2 ( 2015-02), p. 409-418
    In: Inflammatory Bowel Diseases, Oxford University Press (OUP), Vol. 21, No. 2 ( 2015-02), p. 409-418
    Type of Medium: Online Resource
    ISSN: 1078-0998
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2015
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. LB-213-LB-213
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. LB-213-LB-213
    Abstract: Basal-like breast cancers that are negative for clinically important receptors (estrogen, progesterone, and Her2/neu), carry the worst prognosis for patients that are diagnosed with breast cancer. Acheron is a novel protein that was recently demonstrated to be highly expressed in triple-negative basal-like breast cancers. Previous work in our lab has demonstrated that breast cancer cell lines that are engineered to express ectopic Acheron are more invasive, have higher levels of MMP-9 and VEGF, and develop increased tumor burden in vivo in xenografted SCID/beige mice. In order to better understand the role Acheron may play in these basal-like breast cancers, the triple negative breast cancer cell lines MDA 231 and MDA 435 were engineered to express ectopic Acheron. Engineered cells were cultured in serum free media with EGF and FGF to cultivate them as mammary cancer stem-like cells. Relative to empty vector control cells, Acheron-expressing MDA 435 cells demonstrated a greater ability to form acini-like structures in suspension as well as support anchorage independent growth. Cells that were cultivated in basal media with serum were also analyzed for cell adhesion and expression of the laminin adhesion protein CD29 (Beta-I Integrin). Expression of Acheron in either MDA 231 and 435 cells lead to a marked reduction in substrate adhesion on laminin-coated plates relative to controls. Consistent with this observation, Acheron-expressing cells also expressed reduced levels of CD29. FACS analysis for the apoptosis markers Annexin-V binding and propidium iodide exclusion demonstrated that Acheron-expressing cells were 2–3 times less sensitive to the same concentration of the chemotherapeutic drugs: Doxorubicin and docetaxel, and to a lesser extent to ionizing radiation (1.6 fold). In summary, we have extended the analysis of Acheron action in triple-negative basal-like breast cancer cells. We have found that these cells that express ectopic Acheron are more resistant to chemotherapeutic drugs and radiation and have a greater capacity to grow in an anchorage-independent manner. These data support the hypothesis that Acheron expression in triple-negative breast cancers may confer properties that would be anticipated to result in a poorer prognosis. Therefore, Acheron deserves attention and further investigation as a new cancer target in triple-negative basal-like breast cancer. 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 LB-213. doi:10.1158/1538-7445.AM2011-LB-213
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Elsevier BV ; 2012
    In:  Journal of Biological Chemistry Vol. 287, No. 29 ( 2012-07), p. 24821-24831
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 287, No. 29 ( 2012-07), p. 24821-24831
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2012
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4620-4620
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 4620-4620
    Abstract: Recent studies have shown that YKL-40, a secreted glycoprotein, is highly expressed in patients with advanced cancers including breast cancer, colorectal cancer, ovarian cancer, lung cancer, and brain tumors. We have identified YKL-40 as an angiogenic molecule capable of stimulating endothelial cells in different types of cancers. Nevertheless, the therapeutic value of targeting YKL-40 in human cancers remains unknown. Therefore, it was the goal of this study to develop a monoclonal antibody against YKL-40 that could bind specifically and inhibit YKL-40 function with regard to cancer progression. A mouse monoclonal anti-YKL-40 antibody (mAY) was developed using hybridoma technology. mAY could specifically recognize recombinant YKL-40, as well as secreted YKL-40 from the media of two cancerous cell lines, MG63 and U87. Additionally, in functional testing, we demonstrated that mAY inhibited in vitro tube formation on Matrigel of endothelial cells induced by the conditioned media of MG63 and U87 cells. Recombinant YKL-40 treated endothelial cells displayed increased tube formation. mAY significantly abrogated tube formation by blocking recombinant YKL-40. Moreover, mAY reduced YKL-40 activated VEGF receptor 2 (Flk-1/KDR) and intracellular signaling MAP (mitogen-activated protein) kinase extracellular signal-regulated kinase (Erk) 1 and Erk 2. In relation to radiation therapy, treatment of gamma-irradiated U87 cells with mAY induced cell death through a down-regulation of p-AKT and AKT protein expression. Lastly, mAY treatment of U87 xenograft tumors diminished tumor volume, angiogenesis, and progression. These results demonstrate for the first time the therapeutic value of targeting YKL-40 in human cancers which highlights the potential benefits of anti-YKL-40 therapy in patients with a wide spectrum of cancers that over-produce YKL-40. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 4620. doi:1538-7445.AM2012-4620
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 116-116
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly types of cancer, with a 5-year survival of only 10%. One of the major features of PDAC is the dense fibrous stroma, due to the expansion of cancer associated fibroblasts (CAFs) and their extracellular matrix (ECM). This unique environment promotes immunosuppression, lack of nutrients and exclusion or inactivation of antitumor immune cells, representing a challenge for therapies. Recently, we identified the ectopic expression of the neuronal protein Netrin G1 Ligand (NGL-1) in PDAC tissue, including its novel expression in immune cells and CAFs. However, the roles of NGL-1 in the microenvironment of PDAC and in immune cell function are unknown and deserved further investigation. The effects in tumorigenesis were evaluated by orthotopically injecting pancreatic tumor cells in wild-type and NGL-1 full body knockout mice. The roles of NGL-1 in the pro-tumor functions of CAFs were evaluated using our in vitro 3D system, and questioning if NGL-1+ CAFs, compared to NGL-1 knockdown CAFs (CRISPRi), are able to rescue PDAC cell survival under nutrient deprivation, and discern their immunosuppressive profile. For the translational approach, we assessed the overall survival of 140 PDAC patients according to NGL-1 expression in the TME. Myeloid, T and NK cells from tumor-bearing mice tended to overexpress NGL-1 when compared with cells from naïve mice. Moreover, CD8+ and CD4+ T cells from NGL-1 KO mice proliferated more when stimulated in vitro, suggesting that NGL-1 could represent a functional brake for T cells. Moreover, the lack of NGL-1 in stimulated bone marrow-derived macrophages decreased pro-inflammatory cytokine secretion, further suggesting a functional role for NGL-1 in myeloid cells. Of note, NGL-1 KO mice orthotopically injected with PDAC cells developed smaller tumors with decreased secretion of immunosuppressive factors. In accordance, NGL-1 knockdown CAFs did not support PDAC cell survival in vitro and produced less immunosuppressive cytokines, which was phenocopied by the treatment with a peptide targeting NGL-1. Finally, the low expression of NGL-1 in CAFs and immune cells correlated with better survival of PDAC patients, therefore underscoring NGL-1 as a potential new target in PDAC, that could be manipulated in different compartments in pancreatic cancer (cancer cells, CAFs, immune cells). Citation Format: Debora B. Vendramini-Costa, Ralph Francescone, Tiffany Luong, Janusz Franco-Barraza, Igor Astsaturov, Kathy Q. Cai, Andres J. Klein-Szanto, Huamin Wang, Kerry S. Campbell, Edna Cukierman. Stromal expression of the synaptic protein netrin G1 ligand (NGL-1) promotes tumorigenesis and immunosuppression in pancreatic cancer [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 116.
    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
    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) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 3173-3173
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 3173-3173
    Abstract: Pancreatic Ductal Adenocarcinoma (PDAC) is a devastating disease which is in part driven, and supported by changes in its microenvironment, or stroma. These changes occur early, yet PDAC is usually undetected until the disease has metastasized, causing survival rates to drop dramatically. Therefore, there exists an urgency to detect this disease as early as possible and understand its progression. This project dissects the intercellular communication that exists between the primary stromal component, cancer-associated fibroblasts (CAFs), and PDAC cells. Specifically, we focus on how CAF-secreted extracellular vesicles (EVs) promote PDAC progression, with an additional goal to identify biomarkers suitable to generate a non-invasive “liquid biopsy” test for early PDAC detection and prognosis. PDAC communicates with its microenvironment, in part, through the exchange of specific types of EVs, which include exosomes and recently characterized “ectosomes.” We observe distinct types of CAF-derived EVs containing unique surface receptors. One novel surface protein, NetrinG1, is expressed on the plasma membrane of pancreatic CAFs, but not their normal/healthy counterparts. Further, PDAC cells, but not healthy pancreatic epithelial cells, upregulate NetrinG1's lone binding partner, suggesting a role for these factors in PDAC-selective EV uptake. Functional assays designed to test PDAC viability in nutrient deprivation show that CAF-EVs are capable of protecting PDAC cells from the induction of programmed cell death. Further, we show NetrinG1's expression in CAFs is necessary for this EV-mediated survival effect. We also determine that NetrinG1 localizes to the novel “ectosome” EV sub-population, suggesting it possesses unique cargo and is packaged into EVs and secreted through a yet-unknown mechanism separate from canonical exosome trafficking. We also determine, that sub-populations of EVs can be “filtered” locally by the extracellular matrix, based on various EV surface markers. This suggests that the extracellular matrix can play an important role in determining the fate of secreted EVs; which has significant implications for what sub-populations maybe be found circulating systemically in blood, or acting locally in the tumor microenvironment. Pursuing our biomarker goal, we confirm stromal NetrinG1 expression precedes tumorigenesis and are currently seeking to validate the prognostic potential of NetrinG1(+)EVs in blood of PDAC patients. Altogether, this research shines light on a novel mechanism of tumor-stroma communication, and introduces EV biomarkers potentially capable of identifying both early PDAC occurrences and predicted efficacy of certain adjuvant interventions. Citation Format: Kristopher Raghavan, Debora Vendramini, Ralph Francescone, Janusz Franco-Barraza, Edna Cukierman. NetrinG1's pro-tumor role on stroma-derived extracellular vesicles in pancreatic cancer [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 3173.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research Communications, American Association for Cancer Research (AACR), Vol. 2, No. 9 ( 2022-09-19), p. 1017-1036
    Abstract: It is projected that in 5 years, pancreatic cancer will become the second deadliest cancer in the United States. A unique aspect of pancreatic ductal adenocarcinoma (PDAC) is its stroma; rich in cancer-associated fibroblasts (CAFs) and a dense CAF-generated extracellular matrix (ECM). These pathogenic stroma CAF/ECM units cause the collapse of local blood vessels rendering the tumor microenvironment nutrient-poor. PDAC cells are able to survive this state of nutrient stress via support from CAF-secreted material, which includes small extracellular vesicles (sEV). The tumor-supportive CAFs possess a distinct phenotypic profile, compared with normal-like fibroblasts, expressing NetrinG1 (NetG1) at the plasma membrane, and active Integrin α5β1 localized to the multivesicular bodies; traits indicative of poor patient survival. We herein report that NetG1+ CAFs secrete sEVs that stimulate Akt-mediated survival in nutrient-deprived PDAC cells, protecting them from undergoing apoptosis. Furthermore, we show that NetG1 expression in CAFs is required for the prosurvival properties of sEVs. In addition, we report that the above-mentioned CAF markers are secreted in distinct subpopulations of EVs; with NetG1 being enriched in exomeres, and Integrin α5β1 being enriched in exosomes. Finally, we found that NetG1 and Integrin α5β1 were detected in sEVs collected from plasma of patients with PDAC, while their levels were significantly lower in plasma-derived sEVs of sex/age-matched healthy donors. The discovery of these tumor-supporting CAF-EVs elucidates novel avenues in tumor–stroma interactions and pathogenic stroma detection. Significance: Results from this study identified two unique types of tumor-supporting CAF EVs, with evidence of these being detected in patients. Thus, this study facilitates a novel avenue to further dissect the subtleties of the tumor–stroma interactions responsible for PDAC homeostasis and progression, as well as the possibility of establishing future means to detect and monitor dynamic stroma staging.
    Type of Medium: Online Resource
    ISSN: 2767-9764
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 3098144-X
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2014
    In:  The Cancer Journal Vol. 20, No. 3 ( 2014-05), p. 181-189
    In: The Cancer Journal, Ovid Technologies (Wolters Kluwer Health), Vol. 20, No. 3 ( 2014-05), p. 181-189
    Type of Medium: Online Resource
    ISSN: 1528-9117
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2014
    detail.hit.zdb_id: 2126320-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 11, No. 2 ( 2021-02-01), p. 446-479
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) has a poor 5-year survival rate and lacks effective therapeutics. Therefore, it is of paramount importance to identify new targets. Using multiplex data from patient tissue, three-dimensional coculturing in vitro assays, and orthotopic murine models, we identified Netrin G1 (NetG1) as a promoter of PDAC tumorigenesis. We found that NetG1+ cancer-associated fibroblasts (CAF) support PDAC survival, through a NetG1-mediated effect on glutamate/glutamine metabolism. Also, NetG1+ CAFs are intrinsically immunosuppressive and inhibit natural killer cell–mediated killing of tumor cells. These protumor functions are controlled by a signaling circuit downstream of NetG1, which is comprised of AKT/4E-BP1, p38/FRA1, vesicular glutamate transporter 1, and glutamine synthetase. Finally, blocking NetG1 with a neutralizing antibody stunts in vivo tumorigenesis, suggesting NetG1 as potential target in PDAC. Significance: This study demonstrates the feasibility of targeting a fibroblastic protein, NetG1, which can limit PDAC tumorigenesis in vivo by reverting the protumorigenic properties of CAFs. Moreover, inhibition of metabolic proteins in CAFs altered their immunosuppressive capacity, linking metabolism with immunomodulatory function. See related commentary by Sherman, p. 230. This article is highlighted in the In This Issue feature, p. 211
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2607892-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Mitochondrion, Elsevier BV, Vol. 9, No. 1 ( 2009-2), p. 64-
    Type of Medium: Online Resource
    ISSN: 1567-7249
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2009
    detail.hit.zdb_id: 2056923-3
    SSG: 12
    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