Eph receptor B2 (EPHB2) regulates cancer stem cell-like properties in hepatocellular carcinoma.

Q1 Biochemistry, Genetics and Molecular Biology
Stem cell investigation Pub Date : 2022-10-19 eCollection Date: 2022-01-01 DOI:10.21037/sci-2022-031
Jhin Jieh Lim, Edward Kai-Hua Chow, Tan Boon Toh
{"title":"Eph receptor B2 (EPHB2) regulates cancer stem cell-like properties in hepatocellular carcinoma.","authors":"Jhin Jieh Lim, Edward Kai-Hua Chow, Tan Boon Toh","doi":"10.21037/sci-2022-031","DOIUrl":null,"url":null,"abstract":"© Stem Cell Investigation. All rights reserved. Stem Cell Investig 2022;9:5 | https://dx.doi.org/10.21037/sci-2022-031 Hepatocellular carcinoma (HCC) is the most common primary liver cancer with high mortality rates globally. Poor prognosis is associated with eventual treatment resistance and disease relapse, which are often attributed to the presence of a small subset of tumor cells characterized by stem cell-like phenotypes, commonly known as cancer stem cells (CSCs). CSCs possess several key traits such as self-renewing potential, ability to initiate tumor growth, and resistance to conventional therapies targeted at rapidly proliferating cells that form the tumor bulk. In recent years, significant advances have been made in understanding the biology of different tumor CSCs and their corresponding markers, as well as how they interact with the tumor microenvironment. As increasing evidence show that long-term tumor eradication requires eliminating not just the proliferative cells but also the tumor-initiating subpopulations (1,2), greater focus has been placed on developing therapeutic strategies that target CSCs, with potential for future clinical implementation. An important aspect of specifically targeting these tumor-initiating cells rely on the use of cell surface markers. Studies on liver CSCs have uncovered various cell surface antigens such as EpCAM, CD90, CD47, CD13, CD24, and CD133 that identify subpopulations of tumor-initiating cells in HCC (3-8). However, not all cell surface markers are functionally involved in regulating CSC phenotypes in HCC, and specific small molecule inhibitors against them are scarce. In a recent study by Leung et al. (9), the authors identified a novel CSC target for HCC, Eph receptor B2 (EPHB2), that not only acts as a membrane-bound CSC marker but also plays regulatory roles in promoting stemness properties in liver tumor cells. EPHB2 expression was found to increase progressively from normal liver to cirrhotic liver and to HCC in human and mouse models (9). EPHB2 belongs to a large family of Eph receptor tyrosine kinases, which associate with membranebound ephrin ligands through cell-cell contact, leading to bidirectional intracellular signaling and activation of subsequent downstream signaling cascades (10). Eph receptors are involved in various cellular functions and processes, including cellular adhesion, cell migration, vascular development, cell proliferation and survival (10). The complexity of Eph receptor signaling also extends to its involvement in cancer. In fact, the role of EPHB2 in cancer is controversial, as it has been demonstrated to play both tumor-promoting and tumor-suppressive functions depending on the cellular context. In cancers such as glioblastoma, breast cancer, cervical cancer, and cutaneous squamous cell carcinoma, EPHB2 is overexpressed and promotes tumor progression and invasiveness (11-14). In contrast, EPHB2 exerts inhibitory effects on tumor cell migration and invasion, along with its downregulated expression, in cancers of the bladder and colon (15,16). In the context of CSCs, few studies have investigated EPHB2’s role in regulating cancer stemness. EPHB2 was found to be highly expressed in colorectal cancer cells with intestinal stem cell-like phenotypes, and its expression gradually decreased as the cells became differentiated (17,18). In cervical cancer, EPHB2 was involved in orchestrating an epithelial-mesenchymal transition (EMT) program which Editorial Commentary","PeriodicalId":21938,"journal":{"name":"Stem cell investigation","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2022-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/c0/d1/sci-09-2022-031.PMC9618362.pdf","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Stem cell investigation","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.21037/sci-2022-031","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2022/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"Biochemistry, Genetics and Molecular Biology","Score":null,"Total":0}
引用次数: 0

Abstract

© Stem Cell Investigation. All rights reserved. Stem Cell Investig 2022;9:5 | https://dx.doi.org/10.21037/sci-2022-031 Hepatocellular carcinoma (HCC) is the most common primary liver cancer with high mortality rates globally. Poor prognosis is associated with eventual treatment resistance and disease relapse, which are often attributed to the presence of a small subset of tumor cells characterized by stem cell-like phenotypes, commonly known as cancer stem cells (CSCs). CSCs possess several key traits such as self-renewing potential, ability to initiate tumor growth, and resistance to conventional therapies targeted at rapidly proliferating cells that form the tumor bulk. In recent years, significant advances have been made in understanding the biology of different tumor CSCs and their corresponding markers, as well as how they interact with the tumor microenvironment. As increasing evidence show that long-term tumor eradication requires eliminating not just the proliferative cells but also the tumor-initiating subpopulations (1,2), greater focus has been placed on developing therapeutic strategies that target CSCs, with potential for future clinical implementation. An important aspect of specifically targeting these tumor-initiating cells rely on the use of cell surface markers. Studies on liver CSCs have uncovered various cell surface antigens such as EpCAM, CD90, CD47, CD13, CD24, and CD133 that identify subpopulations of tumor-initiating cells in HCC (3-8). However, not all cell surface markers are functionally involved in regulating CSC phenotypes in HCC, and specific small molecule inhibitors against them are scarce. In a recent study by Leung et al. (9), the authors identified a novel CSC target for HCC, Eph receptor B2 (EPHB2), that not only acts as a membrane-bound CSC marker but also plays regulatory roles in promoting stemness properties in liver tumor cells. EPHB2 expression was found to increase progressively from normal liver to cirrhotic liver and to HCC in human and mouse models (9). EPHB2 belongs to a large family of Eph receptor tyrosine kinases, which associate with membranebound ephrin ligands through cell-cell contact, leading to bidirectional intracellular signaling and activation of subsequent downstream signaling cascades (10). Eph receptors are involved in various cellular functions and processes, including cellular adhesion, cell migration, vascular development, cell proliferation and survival (10). The complexity of Eph receptor signaling also extends to its involvement in cancer. In fact, the role of EPHB2 in cancer is controversial, as it has been demonstrated to play both tumor-promoting and tumor-suppressive functions depending on the cellular context. In cancers such as glioblastoma, breast cancer, cervical cancer, and cutaneous squamous cell carcinoma, EPHB2 is overexpressed and promotes tumor progression and invasiveness (11-14). In contrast, EPHB2 exerts inhibitory effects on tumor cell migration and invasion, along with its downregulated expression, in cancers of the bladder and colon (15,16). In the context of CSCs, few studies have investigated EPHB2’s role in regulating cancer stemness. EPHB2 was found to be highly expressed in colorectal cancer cells with intestinal stem cell-like phenotypes, and its expression gradually decreased as the cells became differentiated (17,18). In cervical cancer, EPHB2 was involved in orchestrating an epithelial-mesenchymal transition (EMT) program which Editorial Commentary

Abstract Image

Eph受体B2 (EPHB2)在肝细胞癌中调控肿瘤干细胞样特性。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Stem cell investigation
Stem cell investigation Biochemistry, Genetics and Molecular Biology-Developmental Biology
CiteScore
5.80
自引率
0.00%
发文量
9
期刊介绍: The Stem Cell Investigation (SCI; Stem Cell Investig; Online ISSN: 2313-0792) is a free access, peer-reviewed online journal covering basic, translational, and clinical research on all aspects of stem cells. It publishes original research articles and reviews on embryonic stem cells, induced pluripotent stem cells, adult tissue-specific stem/progenitor cells, cancer stem like cells, stem cell niche, stem cell technology, stem cell based drug discovery, and regenerative medicine. Stem Cell Investigation is indexed in PubMed/PMC since April, 2016.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信