Cargando…

MITF deficiency accelerates GNAQ-driven uveal melanoma

Cutaneous melanoma (CM) and uveal melanoma (UM) both originate from the melanocytic lineage but are primarily driven by distinct oncogenic drivers, BRAF/NRAS or GNAQ/GNA11, respectively. The melanocytic master transcriptional regulator, MITF, is essential for both CM development and maintenance, but...

Descripción completa

Detalles Bibliográficos
Autores principales: Phelps, Grace B., Hagen, Hannah R., Amsterdam, Adam, Lees, Jacqueline A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: National Academy of Sciences 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9172632/
https://www.ncbi.nlm.nih.gov/pubmed/35512098
http://dx.doi.org/10.1073/pnas.2107006119
Descripción
Sumario:Cutaneous melanoma (CM) and uveal melanoma (UM) both originate from the melanocytic lineage but are primarily driven by distinct oncogenic drivers, BRAF/NRAS or GNAQ/GNA11, respectively. The melanocytic master transcriptional regulator, MITF, is essential for both CM development and maintenance, but its role in UM is largely unexplored. Here, we use zebrafish models to dissect the key UM oncogenic signaling events and establish the role of MITF in UM tumors. Using a melanocytic lineage expression system, we showed that patient-derived mutations of GNAQ (GNAQ(Q209L)) or its upstream CYSLTR2 receptor (CYSLTR2(L129Q)) both drive UM when combined with a cooperating mutation, tp53(M214K/M214K). The tumor-initiating potential of the major GNAQ/11 effector pathways, YAP, and phospholipase C-β (PLCβ)–ERK was also investigated in this system and thus showed that while activated YAP (YAP(AA)) induced UM with high potency, the patient-derived PLCβ4 mutation (PLCB4(D630Y)) very rarely yielded UM tumors in the tp53(M214K/M214K) context. Remarkably, mitfa deficiency was profoundly UM promoting, dramatically accelerating the onset and progression of tumors induced by Tg(mitfa:GNAQ(Q209L));tp53(M214K/M214K) or Tg(mitfa:CYSLTR2(L129Q));tp53(M214K/M214K). Moreover, mitfa loss was sufficient to cooperate with GNAQ(Q209L) to drive tp53–wild type UM development and allowed Tg(mitfa:PLCB4(D630Y));tp53(M214K/M214K) melanocyte lineage cells to readily form tumors. Notably, all of the mitfa(−/−) UM tumors, including those arising in Tg(mitfa:PLCB4(D630Y));tp53(M214K/M214K);mitfa(−/−) zebrafish, displayed nuclear YAP while lacking hyperactive ERK indicative of PLCβ signaling. Collectively, these data show that YAP signaling is the major mediator of UM and that MITF acts as a bona fide tumor suppressor in UM in direct opposition to its essential role in CM.