4.8 Article

Zebrafish MITF-Low Melanoma Subtype Models Reveal Transcriptional Subclusters and MITF-Independent Residual Disease

Journal

CANCER RESEARCH
Volume 79, Issue 22, Pages 5769-5784

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/0008-5472.CAN-19-0037

Keywords

-

Categories

Funding

  1. CRUK
  2. University of Edinburgh
  3. Medical Research Council
  4. [PEGASUS]2 Marie Skoodowska-Curie Fellowship [12O5617N]
  5. MRC [MC_UU_00007/16, MC_PC_15075, MC_UU_00007/15, MC_UU_00007/9]
  6. UKRI [MR/R026017/1]
  7. Icelandic Research Fund [184861-051]
  8. University of Iceland
  9. Foundation Against Cancer [2015-143]
  10. European Research Council [ZF-MEL-CHEMBIO-648489]
  11. L'Oreal-Melanoma Research Alliance [401181]
  12. MRC [MC_UU_00007/16, MC_UU_00007/15, MC_UU_12008/1, MC_UU_00009/2, MC_UU_00007/9, MC_UU_00009/1, MR/R026017/1, MC_PC_15075, MC_PC_U127585840] Funding Source: UKRI

Ask authors/readers for more resources

The melanocyte-inducing transcription factor (MITF)-low melanoma transcriptional signature is predictive of poor outcomes for patients, but little is known about its biological significance, and animal models are lacking. Here, we used zebrafish genetic models with low activity of Mitfa (MITF-low) and established that the MITF-low state is causal of melanomaprogression and a predictor of melanoma biological subtype. MITF-low zebrafish melanomas resembled human MITF-low melanomas and were enriched for stem and invasive (mesenchymal) gene signatures. MITF-low activity coupled with a p53 mutation was sufficient to promote superficial growth melanomas, whereas BRAF(V600E) accelerated MITF-low melanoma onset and further promoted the development of MITF-high nodular growth-melanomas. Genetic inhibition of MITF activity led to rapid regression; recurrence occurred following reactivation of MITF. At the regression site, there was minimal residual disease that was resistant to loss of MITF activity (termed MITF-independent cells) with very low-to-no MITF activity or protein. Transcriptomic analysis of MITF-independent residual disease showed enrichment of mesenchymal and neural crest stem cell signatures similar to human therapy-resistant melanomas. Single-cell RNA sequencing revealed MITF-independent residual disease was heterogeneous depending on melanoma subtype. Further, there was a shared subpopulation of residual disease cells that was enriched for a neural crest G(0)-like state that preexisted in the primary tumor and remained present in recurring melanomas. These findings suggest that invasive and stem-like programs coupled with cellular heterogeneity contribute to poor outcomes for MITF-low melanoma patients and that MITF-independent subpopulations are an important therapeutic target to achieve long-term survival outcomes. Significance: This study provides a useful model for MITF-low melanomas and MITF-independent cell populations that can be used to study the mechanisms that drive these tumors as well as identify potential therapeutic options.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available