Egyszerű nézet

dc.contributor.author Fernando, Tharu M
dc.contributor.author Marullo, Rossella
dc.contributor.author Pera Gresely, Benet
dc.contributor.author Phillip, Jude M
dc.contributor.author Yang, Shao Ning
dc.contributor.author Lundell-Smith, Geoffrey
dc.contributor.author Torregroza, Ingrid
dc.contributor.author Ahn, Haelee
dc.contributor.author Evans, Todd
dc.contributor.author Győrffy, Balázs
dc.contributor.author Privé, Gilbert G
dc.contributor.author Hirano, Masayuki
dc.contributor.author Melnick, Ari M
dc.contributor.author Cerchietti, Leandro
dc.date.accessioned 2019-12-07T11:20:34Z
dc.date.available 2019-12-07T11:20:34Z
dc.date.issued 2019
dc.identifier 85065510862
dc.identifier.citation journalVolume=9;journalIssueNumber=5;journalTitle=CANCER DISCOVERY;pagerange=662-679;journalAbbreviatedTitle=CANCER DISCOV;
dc.identifier.uri http://repo.lib.semmelweis.hu//handle/123456789/8027
dc.identifier.uri doi:10.1158/2159-8290.CD-17-1444
dc.description.abstract Several lines of evidence link the canonical oncogene BCL6 to stress response. Here we demonstrate that BCL6 evolved in vertebrates as a component of the HSF1-driven stress response, which has been co-opted by the immune system to support germinal center formation and may have been decisive in the convergent evolution of humoral immunity in jawless and jawed vertebrates. We find that the highly conserved BTB corepressor binding site of BCL6 mediates stress adaptation across vertebrates. We demonstrate that pan-cancer cells hijack this stress tolerance mechanism to aberrantly express BCL6. Targeting the BCL6 BTB domain in cancer cells induces apoptosis and increases susceptibility to repeated doses of cytotoxic therapy. The chemosensitization effect upon BCL6 BTB inhibition is dependent on the derepression of TOX, implicating modulation of DNA repair as a downstream mechanism. Collectively, these data suggest a form of adaptive nononcogene addiction rooted in the natural selection of BCL6 during vertebrate evolution. SIGNIFICANCE: We demonstrate that HSF1 drives BCL6 expression to enable stress tolerance in vertebrates. We identify an HSF1-BCL6-TOX stress axis that is required by cancer cells to tolerate exposure to cytotoxic agents and points toward BCL6-targeted therapy as a way to more effectively kill a wide variety of solid tumors.This article is highlighted in the In This Issue feature, p. 565.
dc.format.extent 662-679
dc.relation.ispartof urn:issn:2159-8274
dc.title BCL6 Evolved to Enable Stress Tolerance in Vertebrates and Is Broadly Required by Cancer Cells to Adapt to Stress
dc.type Journal Article
dc.date.updated 2019-11-27T14:58:00Z
dc.language.rfc3066 en
dc.rights.holder NULL
dc.identifier.mtmt 30757963
dc.identifier.wos 000466758900026
dc.identifier.pubmed 30777872
dc.contributor.department SE/AOK/K/II. Sz. Gyermekgyógyászati Klinika
dc.contributor.institution Semmelweis Egyetem


Kapcsolódó fájlok:

A fájl jelenleg csak egyetemi IP címről érhető el.

Megtekintés/Megnyitás

Ez a rekord az alábbi gyűjteményekben szerepel:

Egyszerű nézet