Identifying in vivo genetic dependencies of melanocyte and melanoma development

  1. Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
  2. Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
  3. Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
  4. Cell and Developmental Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
  5. Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
  6. Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
  7. Department of Genetics, Development and Cell Biology, Iowa State University, IA, USA
  8. Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY, USA
  9. Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, and public reviews.

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Elaine Fuchs
    Howard Hughes Medical Institute, The Rockefeller University, New York, United States of America
  • Senior Editor
    Kathryn Cheah
    University of Hong Kong, Hong Kong, Hong Kong

Reviewer #1 (Public review):

Summary:

Perlee et al. sought to generate a zebrafish line where CRISPR-based gene editing is exclusively limited to the melanocyte lineage, allowing assessment of cell-type restricted gene knockouts. To achieve this, they knocked in Cas9 to the endogenous mitfa locus, as mitfa is a master regulator of melanocyte development. The authors use multiple candidate genes - albino, sox10, tuba1a, ptena/ptenb, tp53 - to demonstrate their system induces lineage-restricted gene editing. This method allows researchers to bypass embryonic lethal and non-cell autonomous phenotypes emerging from whole body knockout (sox10, tuba1a), drive directed phenotypes, such as depigmentation (albino), and induce lineage-specific tumors, such as melanomas (ptena/ptenb, tp53, when accompanied with expression of BRAFV600E). While the genetic approaches are solid, the argued increase in efficiency of this model compared to current tools was untested, and therefore unable to be assessed. Furthermore, the mechanistic explanations proposed to underlie their phenotypes are mostly unfounded, as discussed further in the Weaknesses section. Despite these concerns, there is still a clear use for this genetic methodology and its implementation will be of value to many in vivo researchers.

Strengths:

The strongest component of this manuscript is the genetic control offered by the mitfa:Cas9 system and the ability to make stable, lineage-specific knockouts in zebrafish. This is exemplified by the studies of tuba1a, where the authors nicely show non-cell autonomous mechanisms have obfuscated the role of this gene in melanocyte development. In addition, the mitfa:Cas9 system is elegantly straightforward and can be easily implemented in many labs. Mostly, the figures are clean, controls are appropriate, and phenotypes are reproducible. The invented method is a welcomed addition to the arsenal of genetic tools used in zebrafish.

Weaknesses:

The major weaknesses of the manuscript include the overly bold descriptions of the value of the model and the superficial mechanistic explanations for each biological vignette.

The authors argue that a major advantage of this system is its high efficiency. However, no direct comparison is made with other tools that achieve the same genetic control, such as MAZERATI. This is a missed opportunity to provide researchers the ability to evaluate these two similar genetic approaches. In addition, Fig.1 shows that not all melanocytes express Cas9. This is a major caveat that goes unaddressed. It is of paramount importance to understand the percentage of mitfa+ cells that express Cas9. The histology shown is unclear and too zoomed out of a scale to make any insightful conclusions, especially in Fig.S1. It would also be beneficial to see data regarding Cas9 expression in adult melanocytes, which are distinct from embryonic melanocytes in zebrafish. Moreover, this system still requires the injection of a plasmid encoding gRNAs of interest, which will yield mosaicism. A prime example of this discrepancy is in Fig.6, where sox10 is clearly still present in "sox10 KO" tumors.

The authors argue that their model allows rapid manipulation of melanocyte gene expression. Enthusiasm for the speed of this model is diminished by minimal phenotypes in the F0, as exemplified in Fig.2. Although the authors say >90% of fish have loss of pigmentation, this is misleading as the phenotype is a very weak, partial loss. Only in the F1 generation do robust phenotypes emerge, which takes >6 months to generate. How this is more efficient than other tools that currently exist is unclear and should be discussed in more detail.

In Figure 3, the authors find that melanocyte-specific knockout of sox10 leads to only a 25% reduction in melanocytes in the F1 generation. This is in contradiction to prior literature cited describing sox10 as indispensable for melanocyte development. In addition, the authors argue that sox10 is required for melanocyte regeneration. This claim is not accurate, as >50% of melanocytes killed upon neocuproine treatment can regenerate. This data would indicate that sox10 is required for only a subset of melanocytes to develop (Fig.3C) and for only a subset to regenerate (Fig.3G). This is an interesting finding that is not discussed or interrogated further.

Tumor induction by this model is weak, as indicated by the tumor curves in Figs.5,6. This might be because these fish are mitfa heterozygous. Whereas the avoidance of mitfa overexpression driven by other models including MAZERATI is a benefit of this system, the effect of mitfa heterozygosity on tumor incidence was untested. This is an essential question unaddressed in the manuscript.

In Fig.6, the authors recapitulate previous findings with their model, showing sox10 KO inhibits tumor onset. The tumors that do develop are argued to be highly invasive, have mesenchymal morphology, and undergo phenotypic switching from sox10 to sox9 expression. The data presented do not sufficiently support these claims. The histology is not readily suggestive of invasive, mesenchymal melanomas. Sox10 is still present in many cells and sox9 expression is only found in a small subset (<20%). Whether sox10-null cells are the ones expressing sox9 is untested. If sox9-mediated phenotypic switching is the major driver of these tumors, the authors would need to knockout sox9 and sox10 simultaneously and test whether these "rare" types of tumors still emerge. Additional histological and genetic evaluation is required to make the conclusions presented in Fig.6. It feels like a missed opportunity that the authors did not attempt to study genes of unknown contribution to melanoma with their system.

Overall, this manuscript introduces a solid method to the arsenal of zebrafish genetic tools but falls short of justifying itself as a more efficient and robust approach than what currently exists. The mechanisms provided to explain observed phenotypes are tenuous. Nonetheless, the mitfa:Cas9 approach will certainly be of value to many in vivo biologists and lays the foundation to generate similar methods using other tissue-specific regulators and other Cas proteins.

Reviewer #2 (Public review):

Summary:

This manuscript describes a genetic tool utilizing mutant mitfa-Cas9 expressing zebrafish to knockout genes to analyze their function in melanocytes in a range of assays from developmental biology to tumorigenesis. Overall, the data are convincing and the authors cover potential caveats from their model that might impact its utility for future work.

Strengths:

The authors do an excellent job of characterizing several gene deletions that show the specificity and applicability of the genetic mitfa-Cas9 zebrafish to studying melanocytes.

Weaknesses:

Variability across animals not fully analyzed.

Reviewer #3 (Public review):

Summary:

Perlee et al. present a method for generating cell-type restricted knockouts in zebrafish, focusing on melanocytes. For this method, the authors knock-in a Cas9 encoding sequence into the mitfa locus. This mitfaCas9 line has restricted Cas9 expression, allowing the authors to generate melanocyte-specific knockouts rapidly by follow-up injection of sgRNA expressing transposon vectors.

The paper presents some interesting vignettes to illustrate the utility of their approach. These include 1) a derivation of albino mutant fish as a demonstration of the method's efficiency, 2) an interrogation and novel description of tuba1a as a potential non-autonomous contributor to melanocyte dispersion, and 3) the generation of sox10 deficient melanoma tumors that show "escape" of sox10 loss through upregulation of sox9. The latter two examples highlight the usefulness of cell-type targeted knockouts (Body-wide sox10 and tuba1a loss elicit developmental defects). Additionally, the tumor models involve highly multiplexed sgRNAs for tumor initiation which is nicely facilitated by the stable Cas9.

Strengths:

The approach is clever and could prove very useful for studying melanocytes and other cell types. As the authors hint at in their discussion, this approach would become even more powerful with the generation of other Cas9-restricted lineages so a single sgRNA construct can be screened across many lineages rapidly (or many sgRNA and fish lines screened combinatorially).

The biological findings used to demonstrate the power of the approach are interesting in their own right. If it proves true, tuba1a's non-autonomous effects on melanosome dispersion are striking, and this example demonstrates very nicely how one could use Perlee et al.'s approach to search for other non-autonomous mechanisms systematically. Similarly, the observation of the sox9 escape mechanism with sox10 loss is a beautiful demonstration of the relevance of SOX10/SOX9's reciprocal regulation in vivo. This system would be a very nice model for further interrogating mechanisms/interventions surrounding Sox10 in melanoma.

Finally, the figure presentation is very nice. This work involves complex genetic approaches including multiple fish generations and multiplexed construct injections. The vector diagrams and breeding schemes in the paper make everything very clear/"grok-able," and the paper was enjoyable to read.

Weaknesses:

The mitfa-driven GFP on their sgRNA-expressing cassette is elegant, but it makes one wonder why the endogenous knock-in is necessary. It would strengthen the motivation of the work if the authors could detail the potential advantages and disadvantages of their system compared to expressing Cas9 with a lineage-specific promoter from a transposon in their introduction or discussion.

Related to the above - is mitfa haplosufficient? If the mitfaCas9/+ fish have any notable phenotypes, it would be worth noting for others interested in using this approach to study melanoma and pigmentation.

A core weakness (and also potential strength) of the system is that introduced edits will always be non-clonal (Fig 2H/I). The activity of individual sgRNAs should always be validated in the absence of any noticeable phenotype to interpret a negative result. Additionally, caution should be taken when interpreting results from rare events involving positive outgrowth (like tumorogenesis) to account for the fact many cells in the population might not have biallelic null alleles (i.e., 100% of the gene product removed).

Along those lines: in my opinion, the tuba1a results are the most provocative finding in the paper, but they lack key validation. With respect to cutting activity, the Alt-R and transgenic sgRNA expression approaches are not directly comparable. Since there is no phenotype in the melanocyte specific tuba1a knockouts, the authors must confirm high knockout efficiency with this set of reagents before making the claim there is a non-autonomous phenotype. This can be achieved with GFP+ sorting and NGS like they performed with their albino melanocytes.

The whole-body tuba1a knockout phenotype is expected to be pleiotropic, and this expectation might mask off-target effects. Controls for knockout specificity should be included. For instance, confidence in the claims would greatly increase if the dispersed melanosome phenotype could be recovered with guide-resistant tuba1a re-expression and if melanocyte-restricted tuba1a re-expression failed to rescue. As a less definitive but adequate alternative, the authors could also test if another guide or a morpholino against tuba1a phenocopies the described Alt-R edited fish.

I have similar questions about the sox10 escapers, but these suggestions are less critical for supporting the authors claims (especially given the nice staining). Are the sox10 tumors relatively clonal with respect to sox10 mutations? And are the sox10 tumor mutations mostly biallelic frameshifts or potential missense mutations/single mutations that might not completely remove activity? I am particularly curious as SOX10 doesn't seem to be completely absent (and is still very high in some nuclei) in the immunohistochemistry.

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation