靶向胶质母细胞瘤中原发性纤毛相关信号:药物开发的指导方法。

Yuriy Loskutov, Elena N Pugacheva
{"title":"靶向胶质母细胞瘤中原发性纤毛相关信号:药物开发的指导方法。","authors":"Yuriy Loskutov, Elena N Pugacheva","doi":"10.18632/oncoscience.475","DOIUrl":null,"url":null,"abstract":"Despite unquestionable progress of the recent decades in cancer therapy and anti-cancer drug discovery, the options for glioblastoma (GBM) patients are very limited. Current standard of care includes aggressive surgical resection, radiation therapy, and chemotherapy, however, majority of patients experience recurrence within 32-36 weeks, and median survival is clenching around 10 months. Clearly, there is a need for new strategies to battle GBM. Unfortunately, with GBM we are not on the solid grounds: there is no known common driver mutation to target; the cell lines, commonly used for GBM research, are obsolete, and their origins are in question; even cells GBM arise from, is a topic of harsh debates. Thus, some guidance system would be highly appreciated, a clue towards which signaling cascades are altered and can be targeted. The primary cilium is a ubiquitous, microtubulebased organelle, an important negative regulator of proliferation and a key sensory organelle. Interestingly, in multiple cancer types, cilia tend to be lost, however, the role of this event is not clear. Modeling the loss of cilia in normal cells suggests that it promotes hyperproliferation in Sonic hedgehog (Shh)-independent cells. In the case of Shh-dependent cell populations such as neural stem cells, ablation of primary cilia seems to inhibit their proliferation and lead to hypocellularity [1]. GBM was reported to have a drastically decreased ciliation rate. Only about 10% of GBM cells assemble cilia. Moreover, electron microscopy analysis of GBM biopsies showed that the few cilia, that they do form, possess multiple structural abnormalities [2]. Further inhibition of primary cilia assembly in patientderived GBM primary cell culture through the introduction of a dominant-negative variant of KIF3A seems to have a minimal and non-coherent effect on both cell proliferation in vitro and tumor growth in the mouse intracranial model [3]. On the other hand, even minimal restoration of primary cilia seems to have a tremendous effect on cell proliferation. This clearly suggests that loss of cilia in GBM is a non-random event, and may be associated with the high proliferative capacity of the tumor cells [4]. It also suggests that GBM are developing either from Shh-independent precursor cells or loose Shh-dependency sometime along with the malignant transformation. Thus, our group focused on investigating the impact of primary cilia loss in astrocytes, speculated to be one of the potential GBM precursor [5]. In concordance with previous observations in other cell types, loss of primary cilia promotes proliferation of astrocytes, however it was not sufficient to cause malignant transformation. Interestingly, general growth factor response, measured by ERK1/2 and AKT activation, was significantly changed upon loss of primary cilia. We traced it down to aberrant lysophosphatidic acid (LPA) signaling: the translocation of lysophosphatidic acid receptor 1 (LPAR1) to the plasma membrane in the absence of primary cilia promotes binding the alternative G-protein secondary messengers [6], changing the cellular response to the LPA stimulation. In GBM inhibition of LPA signaling was sufficient to decrease proliferation both in vivo and in vitro, arguing that it is a viable therapeutical approach, which patients might benefit from [7]. LPAR1 is not a unique receptor targeted to primary cilia. A comprehensive list of receptor tyrosine kinases, including PDGFR, EGFR and FGFR, were reported to localize to the cilia [6]. Furthermore, G-protein coupled receptors are very common to be targeted to the cilia, there are at least two independent ciliary targeting sequences within the superfamily [8]. It is clear, that loss of the cilia will effect signaling of at least part of the cascades, in which receptors localize to the cilia. So far, two possible scenarios are confirmed: degradation of the ciliary receptor upon cilia loss (PDGFRαα [6]), and redistribution of the receptor to the plasma membrane (LPAR1 [7]). The systematic studies of the signaling in presence and absence of the cilia will help us to better understand the reasons for decreased ciliation in GBM and other cancer types. At the end, these findings will help us to devise novel targets for anti-cancer therapy. Thus, primary cilium could be the guide pointing us towards successful treatment of GBM.","PeriodicalId":19508,"journal":{"name":"Oncoscience","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2019-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6382257/pdf/","citationCount":"1","resultStr":"{\"title\":\"Targeting primary cilia - associated signaling in glioblastoma: guided approach for drug development.\",\"authors\":\"Yuriy Loskutov, Elena N Pugacheva\",\"doi\":\"10.18632/oncoscience.475\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Despite unquestionable progress of the recent decades in cancer therapy and anti-cancer drug discovery, the options for glioblastoma (GBM) patients are very limited. Current standard of care includes aggressive surgical resection, radiation therapy, and chemotherapy, however, majority of patients experience recurrence within 32-36 weeks, and median survival is clenching around 10 months. Clearly, there is a need for new strategies to battle GBM. Unfortunately, with GBM we are not on the solid grounds: there is no known common driver mutation to target; the cell lines, commonly used for GBM research, are obsolete, and their origins are in question; even cells GBM arise from, is a topic of harsh debates. Thus, some guidance system would be highly appreciated, a clue towards which signaling cascades are altered and can be targeted. The primary cilium is a ubiquitous, microtubulebased organelle, an important negative regulator of proliferation and a key sensory organelle. Interestingly, in multiple cancer types, cilia tend to be lost, however, the role of this event is not clear. Modeling the loss of cilia in normal cells suggests that it promotes hyperproliferation in Sonic hedgehog (Shh)-independent cells. In the case of Shh-dependent cell populations such as neural stem cells, ablation of primary cilia seems to inhibit their proliferation and lead to hypocellularity [1]. GBM was reported to have a drastically decreased ciliation rate. Only about 10% of GBM cells assemble cilia. Moreover, electron microscopy analysis of GBM biopsies showed that the few cilia, that they do form, possess multiple structural abnormalities [2]. Further inhibition of primary cilia assembly in patientderived GBM primary cell culture through the introduction of a dominant-negative variant of KIF3A seems to have a minimal and non-coherent effect on both cell proliferation in vitro and tumor growth in the mouse intracranial model [3]. On the other hand, even minimal restoration of primary cilia seems to have a tremendous effect on cell proliferation. This clearly suggests that loss of cilia in GBM is a non-random event, and may be associated with the high proliferative capacity of the tumor cells [4]. It also suggests that GBM are developing either from Shh-independent precursor cells or loose Shh-dependency sometime along with the malignant transformation. Thus, our group focused on investigating the impact of primary cilia loss in astrocytes, speculated to be one of the potential GBM precursor [5]. In concordance with previous observations in other cell types, loss of primary cilia promotes proliferation of astrocytes, however it was not sufficient to cause malignant transformation. Interestingly, general growth factor response, measured by ERK1/2 and AKT activation, was significantly changed upon loss of primary cilia. We traced it down to aberrant lysophosphatidic acid (LPA) signaling: the translocation of lysophosphatidic acid receptor 1 (LPAR1) to the plasma membrane in the absence of primary cilia promotes binding the alternative G-protein secondary messengers [6], changing the cellular response to the LPA stimulation. In GBM inhibition of LPA signaling was sufficient to decrease proliferation both in vivo and in vitro, arguing that it is a viable therapeutical approach, which patients might benefit from [7]. LPAR1 is not a unique receptor targeted to primary cilia. A comprehensive list of receptor tyrosine kinases, including PDGFR, EGFR and FGFR, were reported to localize to the cilia [6]. Furthermore, G-protein coupled receptors are very common to be targeted to the cilia, there are at least two independent ciliary targeting sequences within the superfamily [8]. It is clear, that loss of the cilia will effect signaling of at least part of the cascades, in which receptors localize to the cilia. So far, two possible scenarios are confirmed: degradation of the ciliary receptor upon cilia loss (PDGFRαα [6]), and redistribution of the receptor to the plasma membrane (LPAR1 [7]). The systematic studies of the signaling in presence and absence of the cilia will help us to better understand the reasons for decreased ciliation in GBM and other cancer types. At the end, these findings will help us to devise novel targets for anti-cancer therapy. Thus, primary cilium could be the guide pointing us towards successful treatment of GBM.\",\"PeriodicalId\":19508,\"journal\":{\"name\":\"Oncoscience\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2019-01-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6382257/pdf/\",\"citationCount\":\"1\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Oncoscience\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.18632/oncoscience.475\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Oncoscience","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.18632/oncoscience.475","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 1
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Targeting primary cilia - associated signaling in glioblastoma: guided approach for drug development.
Despite unquestionable progress of the recent decades in cancer therapy and anti-cancer drug discovery, the options for glioblastoma (GBM) patients are very limited. Current standard of care includes aggressive surgical resection, radiation therapy, and chemotherapy, however, majority of patients experience recurrence within 32-36 weeks, and median survival is clenching around 10 months. Clearly, there is a need for new strategies to battle GBM. Unfortunately, with GBM we are not on the solid grounds: there is no known common driver mutation to target; the cell lines, commonly used for GBM research, are obsolete, and their origins are in question; even cells GBM arise from, is a topic of harsh debates. Thus, some guidance system would be highly appreciated, a clue towards which signaling cascades are altered and can be targeted. The primary cilium is a ubiquitous, microtubulebased organelle, an important negative regulator of proliferation and a key sensory organelle. Interestingly, in multiple cancer types, cilia tend to be lost, however, the role of this event is not clear. Modeling the loss of cilia in normal cells suggests that it promotes hyperproliferation in Sonic hedgehog (Shh)-independent cells. In the case of Shh-dependent cell populations such as neural stem cells, ablation of primary cilia seems to inhibit their proliferation and lead to hypocellularity [1]. GBM was reported to have a drastically decreased ciliation rate. Only about 10% of GBM cells assemble cilia. Moreover, electron microscopy analysis of GBM biopsies showed that the few cilia, that they do form, possess multiple structural abnormalities [2]. Further inhibition of primary cilia assembly in patientderived GBM primary cell culture through the introduction of a dominant-negative variant of KIF3A seems to have a minimal and non-coherent effect on both cell proliferation in vitro and tumor growth in the mouse intracranial model [3]. On the other hand, even minimal restoration of primary cilia seems to have a tremendous effect on cell proliferation. This clearly suggests that loss of cilia in GBM is a non-random event, and may be associated with the high proliferative capacity of the tumor cells [4]. It also suggests that GBM are developing either from Shh-independent precursor cells or loose Shh-dependency sometime along with the malignant transformation. Thus, our group focused on investigating the impact of primary cilia loss in astrocytes, speculated to be one of the potential GBM precursor [5]. In concordance with previous observations in other cell types, loss of primary cilia promotes proliferation of astrocytes, however it was not sufficient to cause malignant transformation. Interestingly, general growth factor response, measured by ERK1/2 and AKT activation, was significantly changed upon loss of primary cilia. We traced it down to aberrant lysophosphatidic acid (LPA) signaling: the translocation of lysophosphatidic acid receptor 1 (LPAR1) to the plasma membrane in the absence of primary cilia promotes binding the alternative G-protein secondary messengers [6], changing the cellular response to the LPA stimulation. In GBM inhibition of LPA signaling was sufficient to decrease proliferation both in vivo and in vitro, arguing that it is a viable therapeutical approach, which patients might benefit from [7]. LPAR1 is not a unique receptor targeted to primary cilia. A comprehensive list of receptor tyrosine kinases, including PDGFR, EGFR and FGFR, were reported to localize to the cilia [6]. Furthermore, G-protein coupled receptors are very common to be targeted to the cilia, there are at least two independent ciliary targeting sequences within the superfamily [8]. It is clear, that loss of the cilia will effect signaling of at least part of the cascades, in which receptors localize to the cilia. So far, two possible scenarios are confirmed: degradation of the ciliary receptor upon cilia loss (PDGFRαα [6]), and redistribution of the receptor to the plasma membrane (LPAR1 [7]). The systematic studies of the signaling in presence and absence of the cilia will help us to better understand the reasons for decreased ciliation in GBM and other cancer types. At the end, these findings will help us to devise novel targets for anti-cancer therapy. Thus, primary cilium could be the guide pointing us towards successful treatment of GBM.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
IPIAD- an augmentation regimen added to standard treatment of pancreatic ductal adenocarcinoma using already-marketed repurposed drugs irbesartan, pyrimethamine, itraconazole, azithromycin, and dapsone IPIAD- an augmentation regimen added to standard treatment of pancreatic ductal adenocarcinoma using already-marketed repurposed drugs irbesartan, pyrimethamine, itraconazole, azithromycin, and dapsone Cancer drug development yesterday, today and tomorrow. Adenoid cystic carcinoma of the head and neck - treatment strategies of a highly malignant tumor with variable localizations. Kinase-targeted therapy in subsets of colorectal cancer.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1