Please use this identifier to cite or link to this item: https://hdl.handle.net/11147/13322
Full metadata record
DC FieldValueLanguage
dc.contributor.authorBarba, Cindy-
dc.contributor.authorEkiz, Hüseyin Atakan-
dc.contributor.authorTang, William Weihao-
dc.contributor.authorGhazaryan, Arevik-
dc.contributor.authorHansen, Mason-
dc.contributor.authorLee, Soh-Hyun-
dc.contributor.authorVoth, Warren Peter-
dc.date.accessioned2023-04-19T12:37:47Z-
dc.date.available2023-04-19T12:37:47Z-
dc.date.issued2023-
dc.identifier.issn2072-6694-
dc.identifier.urihttps://doi.org/10.3390/cancers15051411-
dc.identifier.urihttps://hdl.handle.net/11147/13322-
dc.description.abstractSimple Summary The tumor microenvironment is complex, with interacting immune and tumor cells. Immune cells release inflammatory cytokines, including interferons (IFNs), that drive tumor clearance. However, evidence suggests that tumor cells can also utilize IFNs to enhance growth and survival in certain cases. We demonstrate that interferon gamma (IFN gamma) mediates the metabolic reprogramming of melanoma cells by inducing the essential NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT) gene through STAT1 binding to the NAMPT locus. NAMPT is constitutively expressed in cells during normal homeostasis. However, melanoma cells have higher energetic demands and increased NAMPT. We show that IFN gamma signaling upregulates NAMPT in melanoma cells, increasing cell proliferation and survival. Further, STAT1-inducible Nampt promotes melanoma growth in mice. We provide evidence that melanoma cells directly respond to IFN gamma-activated STAT1 by increasing Nampt, which improves their fitness during tumor immunity. Elucidating mechanisms that regulate NAMPT expression can lead to enhanced therapeutic approaches with immunotherapies that utilize IFN signaling to improve patient outcomes. (1) Background: Immune cells infiltrate the tumor microenvironment and secrete inflammatory cytokines, including interferons (IFNs), to drive antitumor responses and promote tumor clearance. However, recent evidence suggests that sometimes, tumor cells can also harness IFNs to enhance growth and survival. The essential NAD+ salvage pathway enzyme nicotinamide phosphoribosyltransferase (NAMPT) gene is constitutively expressed in cells during normal homeostasis. However, melanoma cells have higher energetic demands and elevated NAMPT expression. We hypothesized that interferon gamma (IFN gamma) regulates NAMPT in tumor cells as a mechanism of resistance that impedes the normal anti-tumorigenic effects of IFN gamma. (2) Methods: Utilizing a variety of melanoma cells, mouse models, Crispr-Cas9, and molecular biology techniques, we explored the importance of IFN gamma-inducible NAMPT during melanoma growth. (3) Results: We demonstrated that IFN gamma mediates the metabolic reprogramming of melanoma cells by inducing Nampt through a Stat1 binding site in the Nampt gene, increasing cell proliferation and survival. Further, IFN/STAT1-inducible Nampt promotes melanoma in vivo. (4) Conclusions: We provided evidence that melanoma cells directly respond to IFN gamma by increasing NAMPT levels, improving their fitness and growth in vivo (control n = 36, SBS KO n = 46). This discovery unveils a possible therapeutic target that may improve the efficacy of immunotherapies involving IFN responses in the clinic.en_US
dc.language.isoenen_US
dc.publisherMDPIen_US
dc.relation.ispartofCancersen_US
dc.rightsinfo:eu-repo/semantics/openAccessen_US
dc.subjectmelanomaen_US
dc.subjectNAMPTen_US
dc.subjectinterferon gammaen_US
dc.subjectNicotinamide Phosphoribosyltransferaseen_US
dc.subjectMetabolismen_US
dc.subjectExpressionen_US
dc.subjectNad(+)en_US
dc.subjectInhibitoren_US
dc.subjectPd-L1en_US
dc.subjectAlphaen_US
dc.subjectBetaen_US
dc.titleInterferon gamma-inducible NAMPT in melanoma cells serves as a mechanism of resistance to enhance tumor growthen_US
dc.typeArticleen_US
dc.institutionauthorEkiz, Hüseyin Atakan-
dc.departmentİzmir Institute of Technology. Molecular Biology and Geneticsen_US
dc.identifier.volume15en_US
dc.identifier.issue5en_US
dc.identifier.wosWOS:000947157100001en_US
dc.identifier.scopus2-s2.0-85149891621en_US
dc.relation.publicationcategoryMakale - Uluslararası Hakemli Dergi - Kurum Öğretim Elemanıen_US
dc.identifier.doi10.3390/cancers15051411-
dc.identifier.pmid36900204en_US
dc.authorscopusid57193688497-
dc.authorscopusid36150568800-
dc.authorscopusid57196482795-
dc.authorscopusid57205656254-
dc.authorscopusid58137587900-
dc.authorscopusid57194563757-
dc.authorscopusid6506737334-
dc.identifier.scopusqualityQ1-
item.grantfulltextopen-
item.openairetypeArticle-
item.fulltextWith Fulltext-
item.cerifentitytypePublications-
item.openairecristypehttp://purl.org/coar/resource_type/c_18cf-
item.languageiso639-1en-
crisitem.author.dept04.03. Department of Molecular Biology and Genetics-
Appears in Collections:Molecular Biology and Genetics / Moleküler Biyoloji ve Genetik
PubMed İndeksli Yayınlar Koleksiyonu / PubMed Indexed Publications Collection
Scopus İndeksli Yayınlar Koleksiyonu / Scopus Indexed Publications Collection
WoS İndeksli Yayınlar Koleksiyonu / WoS Indexed Publications Collection
Files in This Item:
File SizeFormat 
13322.pdf2.28 MBAdobe PDFView/Open
Show simple item record



CORE Recommender

SCOPUSTM   
Citations

2
checked on Apr 5, 2024

WEB OF SCIENCETM
Citations

2
checked on Mar 23, 2024

Page view(s)

58
checked on May 6, 2024

Download(s)

20
checked on May 6, 2024

Google ScholarTM

Check




Altmetric


Items in GCRIS Repository are protected by copyright, with all rights reserved, unless otherwise indicated.