The endosomal pH regulator NHE9 is a driver of stemness in glioblastoma

Author:

Ko Myungjun12ORCID,Makena Monish R1,Schiapparelli Paula2,Suarez-Meade Paola2,Mekile Allatah X1,Lal Bachchu34,Lopez-Bertoni Hernando34ORCID,Kozielski Kristen L56,Green Jordan J5ORCID,Laterra John34,Quiñones-Hinojosa Alfredo2ORCID,Rao Rajini1ORCID

Affiliation:

1. Department of Physiology, The Johns Hopkins University School of Medicine , Baltimore, MD, 21205 , USA

2. Department of Neurosurgery, Mayo Clinic College of Medicine , Jacksonville, FL, 32224 , USA

3. Hugo W. Moser Research Institute at Kennedy Krieger , Baltimore, MD , USA

4. Department of Neurology, The Johns Hopkins University School of Medicine , Baltimore, MD, 21205 , USA

5. Department of Biomedical Engineering, The Johns Hopkins University School of Medicine , Baltimore, MD, 21205 , USA

6. Department of Electrical and Computer Engineering, Technical University of Munich , Munich , Germany

Abstract

Abstract A small population of self-renewing stem cells initiate tumors and maintain therapeutic resistance in glioblastoma (GBM). Given the limited treatment options and dismal prognosis for this disease, there is urgent need to identify drivers of stem cells that could be druggable targets. Previous work showed that the endosomal pH regulator NHE9 is upregulated in GBM and correlates with worse survival prognosis. Here, we probed for aberrant signaling pathways in patient-derived GBM cells and found that NHE9 increases cell surface expression and phosphorylation of multiple receptor tyrosine kinases (RTKs) by promoting their escape from lysosomal degradation. Downstream of NHE9-mediated receptor activation, oncogenic signaling pathways converged on the JAK2-STAT3 transduction axis to induce pluripotency genes Oct4 and Nanog and suppress markers of glial differentiation. We used both genetic and chemical approaches to query the role of endosomal pH in GBM phenotypes. Loss-of-function mutations in NHE9 that failed to alkalinize endosomal lumen did not increase self-renewal capacity of gliomaspheres in vitro. However, monensin, a chemical mimetic of Na+/H+ exchanger activity, and the H+ pump inhibitor bafilomycin bypassed NHE9 to directly alkalinize the endosomal lumen resulting in stabilization of RTKs and induction of Oct4 and Nanog. Using orthotopic models of primary GBM cells we found that NHE9 increased tumor initiation in vivo. We propose that NHE9 initiates inside-out signaling from the endosomal lumen, distinct from the established effects of cytosolic and extracellular pH on tumorigenesis. Endosomal pH may be an attractive therapeutic target that diminishes stemness in GBM, agnostic of specific receptor subtype.

Funder

NIH

United States - Israel Binational Science Foundation

Mayo Clinic Investigator Award

State of Florida Cancer Research Award

Ruth L. Kirschstein Individual National Research Service Award

American Association of Cancer Research-Astra Zeneca Breast Cancer Research Fellowship

HHMI

Gilliam Fellowship for Advanced Study

Publisher

Oxford University Press (OUP)

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