癌症转移中一种新的低氧反应性lncRNA

J. French, S. Edwards
{"title":"癌症转移中一种新的低氧反应性lncRNA","authors":"J. French, S. Edwards","doi":"10.21037/NCRI.2020.03.03","DOIUrl":null,"url":null,"abstract":"© Non-coding RNA Investigation. All rights reserved. Non-coding RNA Investig 2020;4:5 | http://dx.doi.org/10.21037/ncri.2020.03.03 Metastatic breast cancer, refers to the spread of the disease from the breast to other parts of the body, most often to bone, brain, liver or lungs. Despite advances in breast cancer management, most cancer deaths result from metastases that are resistant to systemic therapies (1). Hypoxia (or reduced oxygen availability) is a hallmark of the breast tumor microenvironment and plays an important role in metastatic progression. Breast tumor cells adapt to hypoxia by increasing the activity of the hypoxiainducible transcription factors (HIF1 and HIF2), which regulate the expression of target genes involved in cancer progression (2). Recent studies have implicated long noncoding RNAs (lncRNAs) in hypoxia/HIF-associated breast cancer metastasis, through various mechanisms. Notable examples include the nuclear lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), which is widely reported as a metastasis-promoting lncRNA, however a recent study provided strong evidence that MALAT1 suppresses breast cancer metastasis through inactivation of the TEAD transcription factor (3). HOTAIR (HOX transcript antisense RNA) is highly expressed in primary breast tumors and metastases and associated with poor prognosis. HOTAIR promotes epithelial to mesenchymal transition (EMT) by recruiting the polycomb repressive complex-2 (PRC2) to epigenetically silence target gene promoters (4). Furthermore, hypoxic induction of NEAT1 (nuclear paraspeckle assembly transcript 1) induces the formation of nuclear structures called paraspeckles and retention of F11R (also known as junctional adhesion molecule 1) mRNA in the nucleus (5). Induction of NEAT1 in hypoxia also leads to hallmarks of increased tumorigenesis including acceleration of tumor cell proliferation and inhibition of apoptosis (5). The recent study by Niu et al. (6), provides another example of a hypoxia-responsive lncRNA involved in metastatic breast cancer. The authors initially used RNAseq to identify an hypoxia-inducible antisense lncRNA, called RAB11B-AS1 from MDA-MB-231 breast cancer cells under hypoxic conditions (7). Subsequent ChIP-seq and qPCR showed that HIF2, but not HIF1, was enriched at the RAB11B-AS1 promoter and responsible for hypoxiainduced lncRNA expression. Nui and colleagues then investigated the oncogenic role of RAB11B-AS1 in vitro and in vivo through gainand loss-of function studies. They found ectopic expression of RAB11B-AS1 promoted cell migration and invasion in MDA-MB-231 and SUM159 breast cancer cells, whereas RAB11B-AS1 depletion caused the opposite effect. Orthotopic injection of MDAMB-231 cells, ectopically expressing RAB11B-AS1, into the mammary fat pads of NSG mice, did not affect primary tumor growth, but showed increased expression of the endomucin capillary marker and extensive metastases to the lungs and liver as compared to the control group. Collectively, these results suggest that hypoxia-induced RAB11B-AS1 promotes metastasis of breast cancer cells to distant tissues. To investigate the mechanisms by which RAB11B-AS1 contributes to angiogenesis, the authors analysed RNAseq data from RAB11B-AS1-overexpressed MDA-MB-231 under hypoxia. Gene ontology analysis indicated multiple biological process likely contribute to the phenotype, although it was difficult to rank the results as no value for Editorial Commentary","PeriodicalId":74314,"journal":{"name":"Non-coding RNA investigation","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2020-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.21037/NCRI.2020.03.03","citationCount":"0","resultStr":"{\"title\":\"A new hypoxia-responsive lncRNA in metastatic breast cancer\",\"authors\":\"J. French, S. Edwards\",\"doi\":\"10.21037/NCRI.2020.03.03\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"© Non-coding RNA Investigation. All rights reserved. Non-coding RNA Investig 2020;4:5 | http://dx.doi.org/10.21037/ncri.2020.03.03 Metastatic breast cancer, refers to the spread of the disease from the breast to other parts of the body, most often to bone, brain, liver or lungs. Despite advances in breast cancer management, most cancer deaths result from metastases that are resistant to systemic therapies (1). Hypoxia (or reduced oxygen availability) is a hallmark of the breast tumor microenvironment and plays an important role in metastatic progression. Breast tumor cells adapt to hypoxia by increasing the activity of the hypoxiainducible transcription factors (HIF1 and HIF2), which regulate the expression of target genes involved in cancer progression (2). Recent studies have implicated long noncoding RNAs (lncRNAs) in hypoxia/HIF-associated breast cancer metastasis, through various mechanisms. Notable examples include the nuclear lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), which is widely reported as a metastasis-promoting lncRNA, however a recent study provided strong evidence that MALAT1 suppresses breast cancer metastasis through inactivation of the TEAD transcription factor (3). HOTAIR (HOX transcript antisense RNA) is highly expressed in primary breast tumors and metastases and associated with poor prognosis. HOTAIR promotes epithelial to mesenchymal transition (EMT) by recruiting the polycomb repressive complex-2 (PRC2) to epigenetically silence target gene promoters (4). Furthermore, hypoxic induction of NEAT1 (nuclear paraspeckle assembly transcript 1) induces the formation of nuclear structures called paraspeckles and retention of F11R (also known as junctional adhesion molecule 1) mRNA in the nucleus (5). Induction of NEAT1 in hypoxia also leads to hallmarks of increased tumorigenesis including acceleration of tumor cell proliferation and inhibition of apoptosis (5). The recent study by Niu et al. (6), provides another example of a hypoxia-responsive lncRNA involved in metastatic breast cancer. The authors initially used RNAseq to identify an hypoxia-inducible antisense lncRNA, called RAB11B-AS1 from MDA-MB-231 breast cancer cells under hypoxic conditions (7). Subsequent ChIP-seq and qPCR showed that HIF2, but not HIF1, was enriched at the RAB11B-AS1 promoter and responsible for hypoxiainduced lncRNA expression. Nui and colleagues then investigated the oncogenic role of RAB11B-AS1 in vitro and in vivo through gainand loss-of function studies. They found ectopic expression of RAB11B-AS1 promoted cell migration and invasion in MDA-MB-231 and SUM159 breast cancer cells, whereas RAB11B-AS1 depletion caused the opposite effect. Orthotopic injection of MDAMB-231 cells, ectopically expressing RAB11B-AS1, into the mammary fat pads of NSG mice, did not affect primary tumor growth, but showed increased expression of the endomucin capillary marker and extensive metastases to the lungs and liver as compared to the control group. Collectively, these results suggest that hypoxia-induced RAB11B-AS1 promotes metastasis of breast cancer cells to distant tissues. To investigate the mechanisms by which RAB11B-AS1 contributes to angiogenesis, the authors analysed RNAseq data from RAB11B-AS1-overexpressed MDA-MB-231 under hypoxia. Gene ontology analysis indicated multiple biological process likely contribute to the phenotype, although it was difficult to rank the results as no value for Editorial Commentary\",\"PeriodicalId\":74314,\"journal\":{\"name\":\"Non-coding RNA investigation\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2020-06-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://sci-hub-pdf.com/10.21037/NCRI.2020.03.03\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Non-coding RNA investigation\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.21037/NCRI.2020.03.03\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Non-coding RNA investigation","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.21037/NCRI.2020.03.03","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

©非编码RNA调查。版权所有。非编码RNA研究2020;4:5 | http://dx.doi.org/10.21037/ncri.2020.03.03转移性乳腺癌,是指疾病从乳房扩散到身体其他部位,最常见的是骨骼、大脑、肝脏或肺部。尽管乳腺癌治疗取得了进展,但大多数癌症死亡是由于对全身治疗有抵抗力的转移所致(1)。缺氧(或氧气可用性降低)是乳腺肿瘤微环境的一个标志,在转移进展中起着重要作用。乳腺肿瘤细胞通过增加缺氧诱导转录因子(HIF1和HIF2)的活性来适应缺氧,这些转录因子调节参与癌症进展的靶基因的表达(2)。最近的研究表明,长链非编码rna (lncRNAs)通过各种机制参与缺氧/ hif相关的乳腺癌转移。值得注意的例子包括核lncRNA MALAT1(转移相关肺腺癌转录本1),它被广泛报道为促进转移的lncRNA,但最近的一项研究提供了强有力的证据,表明MALAT1通过使TEAD转录因子失活来抑制乳腺癌转移(3)。HOTAIR (HOX转录反义RNA)在原发性乳腺肿瘤和转移中高表达,并与不良预后相关。HOTAIR通过募集多梳抑制复合体-2 (PRC2)从表观遗传上沉默靶基因启动子,促进上皮细胞向间质转化(EMT)(4)。缺氧诱导NEAT1(核旁斑组装转录本1)诱导核旁斑结构的形成和细胞核中F11R(也称为连接粘附分子1)mRNA的保留(5)。缺氧诱导NEAT1也导致肿瘤发生增加的标志,包括肿瘤细胞增殖加速和细胞凋亡抑制(5)。提供了另一个低氧反应lncRNA参与转移性乳腺癌的例子。作者最初使用RNAseq从缺氧条件下的MDA-MB-231乳腺癌细胞中鉴定出一种低氧诱导的反义lncRNA,称为RAB11B-AS1(7)。随后的ChIP-seq和qPCR显示,在RAB11B-AS1启动子处富集HIF2,而不是HIF1,并负责低氧诱导的lncRNA表达。Nui和同事随后通过功能获得和功能丧失研究RAB11B-AS1在体外和体内的致癌作用。他们发现RAB11B-AS1的异位表达促进了MDA-MB-231和SUM159乳腺癌细胞的细胞迁移和侵袭,而RAB11B-AS1的缺失则产生相反的效果。将异位表达RAB11B-AS1的MDAMB-231细胞原位注射到NSG小鼠的乳腺脂肪垫中,不影响原发肿瘤的生长,但与对照组相比,内膜粘液蛋白毛细血管标志物的表达增加,并广泛转移到肺和肝脏。总之,这些结果表明,缺氧诱导的RAB11B-AS1促进乳腺癌细胞向远处组织转移。为了研究RAB11B-AS1促进血管生成的机制,作者分析了缺氧条件下RAB11B-AS1过表达的MDA-MB-231的RNAseq数据。基因本体论分析表明,多种生物过程可能对表型有贡献,尽管很难对结果进行排序,因为编辑评论没有价值
本文章由计算机程序翻译,如有差异,请以英文原文为准。
A new hypoxia-responsive lncRNA in metastatic breast cancer
© Non-coding RNA Investigation. All rights reserved. Non-coding RNA Investig 2020;4:5 | http://dx.doi.org/10.21037/ncri.2020.03.03 Metastatic breast cancer, refers to the spread of the disease from the breast to other parts of the body, most often to bone, brain, liver or lungs. Despite advances in breast cancer management, most cancer deaths result from metastases that are resistant to systemic therapies (1). Hypoxia (or reduced oxygen availability) is a hallmark of the breast tumor microenvironment and plays an important role in metastatic progression. Breast tumor cells adapt to hypoxia by increasing the activity of the hypoxiainducible transcription factors (HIF1 and HIF2), which regulate the expression of target genes involved in cancer progression (2). Recent studies have implicated long noncoding RNAs (lncRNAs) in hypoxia/HIF-associated breast cancer metastasis, through various mechanisms. Notable examples include the nuclear lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1), which is widely reported as a metastasis-promoting lncRNA, however a recent study provided strong evidence that MALAT1 suppresses breast cancer metastasis through inactivation of the TEAD transcription factor (3). HOTAIR (HOX transcript antisense RNA) is highly expressed in primary breast tumors and metastases and associated with poor prognosis. HOTAIR promotes epithelial to mesenchymal transition (EMT) by recruiting the polycomb repressive complex-2 (PRC2) to epigenetically silence target gene promoters (4). Furthermore, hypoxic induction of NEAT1 (nuclear paraspeckle assembly transcript 1) induces the formation of nuclear structures called paraspeckles and retention of F11R (also known as junctional adhesion molecule 1) mRNA in the nucleus (5). Induction of NEAT1 in hypoxia also leads to hallmarks of increased tumorigenesis including acceleration of tumor cell proliferation and inhibition of apoptosis (5). The recent study by Niu et al. (6), provides another example of a hypoxia-responsive lncRNA involved in metastatic breast cancer. The authors initially used RNAseq to identify an hypoxia-inducible antisense lncRNA, called RAB11B-AS1 from MDA-MB-231 breast cancer cells under hypoxic conditions (7). Subsequent ChIP-seq and qPCR showed that HIF2, but not HIF1, was enriched at the RAB11B-AS1 promoter and responsible for hypoxiainduced lncRNA expression. Nui and colleagues then investigated the oncogenic role of RAB11B-AS1 in vitro and in vivo through gainand loss-of function studies. They found ectopic expression of RAB11B-AS1 promoted cell migration and invasion in MDA-MB-231 and SUM159 breast cancer cells, whereas RAB11B-AS1 depletion caused the opposite effect. Orthotopic injection of MDAMB-231 cells, ectopically expressing RAB11B-AS1, into the mammary fat pads of NSG mice, did not affect primary tumor growth, but showed increased expression of the endomucin capillary marker and extensive metastases to the lungs and liver as compared to the control group. Collectively, these results suggest that hypoxia-induced RAB11B-AS1 promotes metastasis of breast cancer cells to distant tissues. To investigate the mechanisms by which RAB11B-AS1 contributes to angiogenesis, the authors analysed RNAseq data from RAB11B-AS1-overexpressed MDA-MB-231 under hypoxia. Gene ontology analysis indicated multiple biological process likely contribute to the phenotype, although it was difficult to rank the results as no value for Editorial Commentary
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
×
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