Back to Search Start Over

An immature, dedifferentiated, and lineage-deconstrained cone precursor origin of N-Myc-initiated retinoblastoma.

Authors :
Singh HP
Shayler DWH
Fernandez GE
Thornton ME
Craft CM
Grubbs BH
Cobrinik D
Source :
Proceedings of the National Academy of Sciences of the United States of America [Proc Natl Acad Sci U S A] 2022 Jul 12; Vol. 119 (28), pp. e2200721119. Date of Electronic Publication: 2022 Jul 08.
Publication Year :
2022

Abstract

Most retinoblastomas develop from maturing cone precursors in response to biallelic RB1 loss and are dependent on cone maturation-related signaling. Additionally, ∼2% lack RB1 mutations but have MYCN amplification ( MYCN <superscript>A</superscript> ), N-Myc protein overexpression, and more rapid and invasive growth, yet the MYCN <superscript>A</superscript> retinoblastoma cell of origin and basis for its responses to deregulated N-Myc are unknown. Here, using explanted cultured retinae, we show that ectopic N-Myc induces cell cycle entry in cells expressing markers of several retinal types yet induces continuous proliferation and tumorigenesis only in cone precursors. Unlike the response to RB1 loss, both immature cone arrestin-negative (ARR3 <superscript>-</superscript> ) and maturing ARR3 <superscript>+</superscript> cone precursors proliferate, and maturing cone precursors rapidly dedifferentiate, losing ARR3 as well as L/M-opsin expression. N-Myc-overexpressing retinal cells also lose cell lineage constraints, occasionally coexpressing the cone-specific RXRγ with the rod-specific NRL or amacrine-specific AP2α and widely coexpressing RXRγ with the progenitor and Müller cell-specific SOX9 and retinal ganglion cell-specific BRN3 and GAP43. Mechanistically, N-Myc induced Cyclin D2 and CDK4 overexpression, pRB phosphorylation, and SOX9-dependent proliferation without a retinoma-like stage that characterizes pRB-deficient retinoblastoma, despite continuous p16 <superscript>INK4A</superscript> expression. Orthotopic xenografts of N-Myc-overexpressing retinal cells formed tumors with retinal cell marker expression similar to those in MYCN -transduced retinae and MYCN <superscript>A</superscript> retinoblastomas in patients. These findings demonstrate the MYCN <superscript>A</superscript> retinoblastoma origin from immature and lineage-deconstrained cone precursors, reveal their opportunistic use of an undifferentiated retinal progenitor cell feature, and illustrate that different cancer-initiating mutations cooperate with distinct developmental stage-specific cell signaling circuitries to drive retinoblastoma tumorigenesis.

Details

Language :
English
ISSN :
1091-6490
Volume :
119
Issue :
28
Database :
MEDLINE
Journal :
Proceedings of the National Academy of Sciences of the United States of America
Publication Type :
Academic Journal
Accession number :
35867756
Full Text :
https://doi.org/10.1073/pnas.2200721119