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
Filter
  • Yue, Ming  (4)
  • 1
    In: Molecular Cell, Elsevier BV, Vol. 64, No. 3 ( 2016-11), p. 493-506
    Type of Medium: Online Resource
    ISSN: 1097-2765
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 2001948-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: The Journal of Pathology, Wiley, Vol. 235, No. 1 ( 2015-01), p. 90-100
    Abstract: Amplification of the MYCN gene in human neuroblastoma predicts poor prognosis and resistance to therapy. We previously showed that MYCN ‐amplified neuroblastoma cells constantly require large amounts of glutamine to support their unabated growth. However, the identity and regulation of the transporter(s) that capture glutamine in MYCN ‐amplified neuroblastoma cells and the clinical significance of the transporter(s) in neuroblastoma diagnosis remain largely unknown. Here, we performed a systemic glutamine influx analysis and identified that MYCN ‐amplified neuroblastoma cells predominantly rely on activation of ASCT2 (solute carrier family 1 member 5, SLC1A5 ) to maintain sufficient levels of glutamine essential for the TCA cycle anaplerosis. Consequently, ASCT2 depletion profoundly inhibited glutaminolysis, concomitant with a substantial decrease in cell proliferation and viability in vitro and inhibition of tumourigenesis in vivo . Mechanistically, we identified ATF4 as a novel regulator which coordinates with N‐Myc to directly activate ASCT2 expression. Of note, ASCT2 expression, which correlates with that of N‐Myc and ATF4 , is markedly elevated in high‐stage neuroblastoma tumour samples compared with low‐stage ones. More importantly, high ASCT2 expression is significantly associated with poor prognosis and survival of neuroblastoma patients. In aggregate, these findings elucidate a novel mechanism depicting how cell autonomous insults ( MYCN amplification) and microenvironmental stresses ( ATF4 induction) in concert coordinate ASCT2 activation to promote aggressive neuroblastoma progression, and establish ASCT2 as a novel biomarker in patient prognosis and stratification. Copyright © 2014 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd
    Type of Medium: Online Resource
    ISSN: 0022-3417 , 1096-9896
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2015
    detail.hit.zdb_id: 1475280-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1134-1134
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1134-1134
    Abstract: Elevated glutamine metabolism is an essential feature for malignant transformation. Glutaminase-catalyzed deamidation of glutamine to glutamate is a key step in both glutaminolysis and synthesis of ROS-scavenging glutathione. Two predominant human isozymes of glutaminase, kidney-type GLS1 (also called GLS) and liver-type GLS2, are involved in regulation of glutaminolysis. It has been shown the Myc family member, c-Myc, specifically potentiates elevated expression of GLS1 and promotes the downstream utilization of glutamine carbon in anaplerosis in liver cancer, whereas p53 tumor suppressor specifically activates GLS2 to support cellular defense against oxidative stress, suggesting GLS1 promoted tumorigenesis whereas GLS2 may inhibited it. However, whether cancer cells of different origins use a common or unique signal pathways to regulate glutamine metabolism remains largely unexplored. Using MYCN-amplified neuroblastoma as a model system, we identified GLS2 expression dramatically increased while that of GLS1 substantially decreased in the tumorigenic process of N-Myc transformed neuroblastoma. GLS2 depletion profoundly inhibited glutaminolysis concomitant with a substantial decrease in cell proliferation and viability in vitro and inhibition of tumorigenesis in vivo. Mechanistically, N-Myc, but not p53, selectively activates GLS2 transcription by direct binding to a conserved Myc-binding site within GLS2 promoter. In support of this notion, we found significant elevation of GLS2 expression concomitant with considerable GLS1 down-regulation in primary tumors obtained from MYCN-amplified neuroblastoma patients (invariably with poor prognosis) in comparison with those from MYCN non-amplified patients. In aggregate, these studies suggest that the metabolic regulation of c-Myc induced liver tumors differs from that of N-Myc induced neuroblastoma tumors which is likely to depend on both the genotype and the tissues of origin and have implications regarding the design of selective therapies targeting tumor metabolism. Citation Format: Daibiao Xiao, Ping Ren, Hexiu Su, Ming Yue, Ruijuan Xiu, Lei Gan, Hudan Liu, Guoliang Qing. N-Myc promotes glutamine anaplerosis and aggressive tumor progression through direct GLS2 activation. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1134. doi:10.1158/1538-7445.AM2015-1134
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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
    Impact Journals, LLC ; 2015
    In:  Oncotarget Vol. 6, No. 38 ( 2015-12-01), p. 40655-40666
    In: Oncotarget, Impact Journals, LLC, Vol. 6, No. 38 ( 2015-12-01), p. 40655-40666
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2560162-3
    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...