Your browser doesn't support javascript.
loading
An immature, dedifferentiated, and lineage-deconstrained cone precursor origin of N-Myc-initiated retinoblastoma.
Singh, Hardeep P; Shayler, Dominic W H; Fernandez, G Esteban; Thornton, Matthew E; Craft, Cheryl Mae; Grubbs, Brendan H; Cobrinik, David.
Afiliación
  • Singh HP; The Vision Center, Children's Hospital Los Angeles, Los Angeles, CA 90027.
  • Shayler DWH; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027.
  • Fernandez GE; USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033.
  • Thornton ME; The Vision Center, Children's Hospital Los Angeles, Los Angeles, CA 90027.
  • Craft CM; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027.
  • Grubbs BH; The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027.
  • Cobrinik D; Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033.
Proc Natl Acad Sci U S A ; 119(28): e2200721119, 2022 07 12.
Article en En | MEDLINE | ID: mdl-35867756
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 (MYCNA), N-Myc protein overexpression, and more rapid and invasive growth, yet the MYCNA 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-) and maturing ARR3+ 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 p16INK4A expression. Orthotopic xenografts of N-Myc-overexpressing retinal cells formed tumors with retinal cell marker expression similar to those in MYCN-transduced retinae and MYCNA retinoblastomas in patients. These findings demonstrate the MYCNA 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.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Retinoblastoma / Células Fotorreceptoras Retinianas Conos / Neoplasias de la Retina / Carcinogénesis / Proteína Proto-Oncogénica N-Myc Límite: Humans Idioma: En Revista: Proc Natl Acad Sci U S A Año: 2022 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Retinoblastoma / Células Fotorreceptoras Retinianas Conos / Neoplasias de la Retina / Carcinogénesis / Proteína Proto-Oncogénica N-Myc Límite: Humans Idioma: En Revista: Proc Natl Acad Sci U S A Año: 2022 Tipo del documento: Article