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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2685-2685
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 2685-2685
    Abstract: Of the four molecular subtypes of melanoma (BRAF, NRAS, NF1, and triple WT), NRAS mutant tumors are notoriously more aggressive and challenging to treat. The frequent occurrence of NRAS mutations in benign, congenital nevi suggests that these genetic alterations are early events in melanoma genesis. Since NRAS mutant nevi can remain indolent for years, or even a lifetime, secondary genetic events must be required to drive melanoma initiation. Ultraviolet (UV) light, a major risk factor for melanoma, is known to directly damage DNA and alter the skin microenvironment. Therefore, we hypothesized that the cell-extrinsic and intrinsic events triggered by UV exposure would cooperate with NrasQ61R mutations to drive melanoma formation in vivo. To test this hypothesis we employed a genetically engineered mouse model (i.e. TpN61R) that combines CRE-inducible expression of NrasQ61R with the loss of p16INK4a in melanocytes. Following neonatal CRE induction, we exposed TpN61R mice to a single, nonerythemic dose of UVB radiation and monitored them for the development of melanoma. Exposure to UVB radiation dramatically reduced the melanoma-free survival of TpN61R mice by 80% and increased tumor multiplicity (average 1.2 to 3.4 tumors/mouse); however, it had no impact on tumor growth rates, overall skin proliferation, immune infiltration, or vascularity. To test the respective roles of NrasQ61R and p16INK4a loss in UV-induced melanoma genesis, we generated mice with either melanocytic NrasQ61R expression or p16INK4a loss. Mice with p16INK4a loss alone did not develop tumors in the presence or absence of UV. By contrast, in UV treated TN61R mice, NrasQ61R was sufficient to initiate melanoma formation, albeit with a 66% longer latency than UV exposed TpN61R animals (9.14wks vs. 5.5wks). To determine if NrasQ61R mutations had to be present at the time of UV exposure to drive early melanoma formation, TpN61R mice were exposed to UV prior to CRE activation. Even when NrasQ61R expression was induced three days after UV exposure, melanoma formation was rapidly accelerated. Therefore, NRAS mutations do not need to be present at time of UV exposure to promote early melanoma formation. Genomic analyses comparing spontaneous and UV-induced TpN61R melanomas failed to uncover common secondary mutations that explain the exquisite cooperativity between NrasQ61R and UV light in driving melanoma formation. For this reason, we sought to identify UV-induced, cell-extrinsic factors that might facilitate the initiation of NrasQ61R mutant melanomas via cytokine profiling. Through these analyses we identified UV-induced pro-inflammatory cytokines that could be therapeutically targeted to limit the initiation of NRAS-mutant melanomas. Together, this work explains why UV exposure is such a significant risk factor for melanoma and provides original mechanistic insight into how this deadly disease might be prevented. Citation Format: Rebecca C. Hennessey, Andrea M. Holderbaum, James E. Gillahan, Conor Delaney, Xing Tang, Raleigh Kladney, Christin E. Burd. Ultraviolet light cooperates with NrasQ61R mutations to drive malignant melanoma. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 2685.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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. 76, No. 14_Supplement ( 2016-07-15), p. 900-900
    Abstract: Cutaneous melanoma claims the life of one American every hour, and while the etiology of this tumor type is not entirely understood, exposure to ultraviolet (UV) radiation (280-340nm) is a major risk factor. For this reason, the use of UV-blocking sunscreens is strongly advocated; however, few studies have tested the relative efficacy of these agents in preventing melanoma formation in vivo. Here, we employed a new genetically engineered mouse model (TpN61R) to examine the ability of 6 chemically distinct SPF30 sunscreens to prevent melanoma. In this TpN61R model, topical 4-hydroxytamoxifen (4OHT) treatment induces the melanocyte-specific expression of oncogenic NRas as well as inactivation of the p16INK4a tumor suppressor. These genetic lesions co-occur in ∼24% of all human melanomas, making the TpN61R model biologically relevant. Since NRAS mutations are an early and UV-independent event in human melanoma, TpN61R mice were first painted with 4OHT and then exposed to a single dose of 4.5 kJ/m2 UVB light one day later. Exposed skin from TpN61R mice showed transient cyclobutane pyrimidine dimer (CPD) formation, however no evidence of edema or inflammation was observed. Despite these mild effects, UVB exposure reduced the melanoma-free survival of TpN61R mice by 80% and increased tumor incidence rate from 1.2 to 3.4 tumors/mouse. Further experiments using a variety of UVB doses (0.25, 1.0, 2.3 and 9.0 kJ/m2) in the TpN61R model revealed a dose-dependent increase in early melanoma incidence. Together, these data establish the exquisite cooperation of UV light and oncogenic NRas mutations in driving melanoma. Taking advantage of this unique model, we tested the preventative efficacy of SPF30 sunscreens with differing chemical composition. Sunscreens components included UVA (avobenzene), UVB (homosalate, octisalate) and broad spectrum (oxybenzone, octocrylene, zinc oxide) blocking agents. Application of sunscreen prior to UVB exposure decreased DNA damage, delayed melanoma onset and reduced tumor incidence in a sunscreen-dependent manner. Thus, SPF30 sunscreens do not equally prevent UVB-induced, NRAS mutant melanoma. This work establishes the first in vivo system to test sunscreen efficacy in NRAS-driven melanoma and will help direct the development of improved melanoma preventatives. Citation Format: Andrea M. Holderbaum, Rebecca C. Hennessey, James E. Gillahan, Anamaria Bonilla, Conor Delaney, Raleigh D. Kladney, Kathleen L. Tober, Tatiana M. Oberyszyn, Christin E. Burd. In vivo modeling of NRAS-mutant melanoma reveals differential preventative efficacy amongst SPF30 sunscreens. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 900.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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: Pigment Cell & Melanoma Research, Wiley, Vol. 30, No. 5 ( 2017-09), p. 477-487
    Abstract: To mitigate melanoma risk, sunscreen use is widely advocated; yet, the ability of sunscreens to prevent melanoma remains controversial. Here, we test the tenet that sunscreens limit melanoma risk by blocking ultraviolet radiation ( UV )‐induced DNA damage using murine models that recapitulate the genetics and spontaneous evolution of human melanoma. We find that a single, non‐erythematous dose of UV dramatically accelerates melanoma onset and increases tumor multiplicity in mice carrying an endogenous, melanocyte‐specific NR as 61R allele. By contrast, transient UV exposure does not alter tumor onset in mice lacking p16 INK 4a or harboring an NR as 12D allele. To block the rapid onset of melanoma cooperatively caused by UV and NR as 61R , we employed a variety of aerosol sunscreens. While all sunscreens delayed melanoma formation and blocked UV ‐induced DNA damage, differences in aerosol output (i.e., amount applied/cm 2 ) caused variability in the cancer preventative efficacy of products with identical sunburn protection factor ( SPF ) ratings.
    Type of Medium: Online Resource
    ISSN: 1755-1471 , 1755-148X
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2425880-5
    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...