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

Author:

Singh Hardeep P.123ORCID,Shayler Dominic W. H.12,Fernandez G. Esteban2ORCID,Thornton Matthew E.4ORCID,Craft Cheryl Mae35ORCID,Grubbs Brendan H.4,Cobrinik David12367ORCID

Affiliation:

1. The Vision Center, Children‘s Hospital Los Angeles, Los Angeles, CA 90027

2. The Saban Research Institute, Children‘s Hospital Los Angeles, Los Angeles, CA 90027

3. USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033

4. Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033

5. Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033

6. Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033

7. Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033

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 A ), N-Myc protein overexpression, and more rapid and invasive growth, yet the MYCN A 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 p16 INK4A 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 A retinoblastomas in patients. These findings demonstrate the MYCN A 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.

Funder

HHS | NIH | Center for Scientific Review

Publisher

Proceedings of the National Academy of Sciences

Subject

Multidisciplinary

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3