Articles

< Previous         Next >  
Impaired dNKAP function drives genome instability and tumorigenic growth in Drosophila epithelia
Ting Guo1,2,3,† , Chen Miao1,2,3,† , Zhonghua Liu4,† , Jingwei Duan1,2,3 , Yanbin Ma1,2,3 , Xiao Zhang1,2 , Weiwei Yang1,2 , Maoguang Xue1,2 , Qiannan Deng1,2 , Pengfei Guo1,2 , Yongmei Xi1,2 , Xiaohang Yang1,2 , Xun Huang4 , Wanzhong Ge1,2,3,5,*
1Division of Human Reproduction and Developmental Genetics, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
2Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310058, China
3Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
4State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
5Cancer Center, Zhejiang University, Hangzhou 310058, China
These authors contributed equally to this work.
*Correspondence to:Wanzhong Ge , Email:wanzhongge@zju.edu.cn
J Mol Cell Biol, Volume 15, Issue 12, December 2023, mjad078,  https://doi.org/10.1093/jmcb/mjad078
Keyword: Drosophila wing imaginal disc, dNKAP, genome instability, tumorigenic growth

Mutations or dysregulated expression of NF-kappaB-activating protein (NKAP) family genes have been found in human cancers. How NKAP family gene mutations promote tumor initiation and progression remains to be determined. Here, we characterized dNKAP, the Drosophila homolog of NKAP, and showed that impaired dNKAP function causes genome instability and tumorigenic growth in a Drosophila epithelial tumor model. dNKAP-knockdown wing imaginal discs exhibit tumorigenic characteristics, including tissue overgrowth, cell-invasive behavior, abnormal cell polarity, and cell adhesion defects. dNKAP knockdown causes both R-loop accumulation and DNA damage, indicating the disruption of genome integrity. Further analysis showed that dNKAP knockdown induces c-Jun N-terminal kinase (JNK)-dependent apoptosis and causes aberrant cell proliferation in distinct cell populations. Activation of the Notch and JAK/STAT signaling pathways contributes to the tumorigenic growth of dNKAP-knockdown tissues. Furthermore, JNK signaling is essential for dNKAP depletion-mediated cell invasion. Transcriptome analysis of dNKAP-knockdown tissues confirmed the misregulation of signaling pathways involved in promoting tumorigenesis and revealed abnormal regulation of metabolic pathways. dNKAP knockdown and oncogenic Ras, Notch, or Yki mutations show synergies in driving tumorigenesis, further supporting the tumor-suppressive role of dNKAP. In summary, this study demonstrates that dNKAP plays a tumor-suppressive role by preventing genome instability in Drosophila epithelia and thus provides novel insights into the roles of human NKAP family genes in tumor initiation and progression.