缺乏天冬酰胺合成酶的 HCT116 异种移植物的生长特征因性别而异。

IF 3.8 3区 医学 Q2 GENETICS & HEREDITY Human Genomics Pub Date : 2024-06-17 DOI:10.1186/s40246-024-00635-3
Oladimeji Aladelokun, Lingeng Lu, Jie Zheng, Hong Yan, Abhishek Jain, Joanna Gibson, Sajid A Khan, Caroline H Johnson
{"title":"缺乏天冬酰胺合成酶的 HCT116 异种移植物的生长特征因性别而异。","authors":"Oladimeji Aladelokun, Lingeng Lu, Jie Zheng, Hong Yan, Abhishek Jain, Joanna Gibson, Sajid A Khan, Caroline H Johnson","doi":"10.1186/s40246-024-00635-3","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Sex-related differences in colorectal (CRC) incidence and mortality are well-documented. However, the impact of sex on metabolic pathways that drive cancer growth is not well understood. High expression of asparagine synthetase (ASNS) is associated with inferior survival for female CRC patients only. Here, we used a CRISPR/Cas9 technology to generate HCT116 ASNS<sup>-/-</sup> and HCT 116 ASNS<sup>+/+</sup> cancer cell lines. We examine the effects of ASNS deletion on tumor growth and the subsequent rewiring of metabolic pathways in male and female Rag2/IL2RG mice.</p><p><strong>Results: </strong>ASNS loss reduces cancer burden in male and female tumor-bearing mice (40% reduction, q < 0.05), triggers metabolic reprogramming including gluconeogenesis, but confers a survival improvement (30 days median survival, q < 0.05) in female tumor-bearing mice alone. Transcriptomic analyses revealed upregulation of G-protein coupled estrogen receptor (GPER1) in tumors from male and female mice with HCT116 ASNS<sup>-/-</sup> xenograft. Estradiol activates GPER1 in vitro in the presence of ASNS and suppresses tumor growth.</p><p><strong>Conclusions: </strong>Our study indicates that inferior survival for female CRC patients with high ASNS may be due to metabolic reprogramming that sustains tumor growth. These findings have translational relevance as ASNS/GPER1 signaling could be a future therapeutic target to improve the survival of female CRC patients.</p>","PeriodicalId":13183,"journal":{"name":"Human Genomics","volume":null,"pages":null},"PeriodicalIF":3.8000,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11184737/pdf/","citationCount":"0","resultStr":"{\"title\":\"Growth characteristics of HCT116 xenografts lacking asparagine synthetase vary according to sex.\",\"authors\":\"Oladimeji Aladelokun, Lingeng Lu, Jie Zheng, Hong Yan, Abhishek Jain, Joanna Gibson, Sajid A Khan, Caroline H Johnson\",\"doi\":\"10.1186/s40246-024-00635-3\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Sex-related differences in colorectal (CRC) incidence and mortality are well-documented. However, the impact of sex on metabolic pathways that drive cancer growth is not well understood. High expression of asparagine synthetase (ASNS) is associated with inferior survival for female CRC patients only. Here, we used a CRISPR/Cas9 technology to generate HCT116 ASNS<sup>-/-</sup> and HCT 116 ASNS<sup>+/+</sup> cancer cell lines. We examine the effects of ASNS deletion on tumor growth and the subsequent rewiring of metabolic pathways in male and female Rag2/IL2RG mice.</p><p><strong>Results: </strong>ASNS loss reduces cancer burden in male and female tumor-bearing mice (40% reduction, q < 0.05), triggers metabolic reprogramming including gluconeogenesis, but confers a survival improvement (30 days median survival, q < 0.05) in female tumor-bearing mice alone. Transcriptomic analyses revealed upregulation of G-protein coupled estrogen receptor (GPER1) in tumors from male and female mice with HCT116 ASNS<sup>-/-</sup> xenograft. Estradiol activates GPER1 in vitro in the presence of ASNS and suppresses tumor growth.</p><p><strong>Conclusions: </strong>Our study indicates that inferior survival for female CRC patients with high ASNS may be due to metabolic reprogramming that sustains tumor growth. These findings have translational relevance as ASNS/GPER1 signaling could be a future therapeutic target to improve the survival of female CRC patients.</p>\",\"PeriodicalId\":13183,\"journal\":{\"name\":\"Human Genomics\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.8000,\"publicationDate\":\"2024-06-17\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11184737/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Human Genomics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s40246-024-00635-3\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"GENETICS & HEREDITY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Human Genomics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s40246-024-00635-3","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

摘要

背景:结直肠癌(CRC)发病率和死亡率的性别差异已得到充分证实。然而,人们对性别对驱动癌症生长的代谢途径的影响还不甚了解。天冬酰胺合成酶(ASNS)的高表达仅与女性 CRC 患者的低生存率有关。在这里,我们使用 CRISPR/Cas9 技术生成了 HCT116 ASNS-/- 和 HCT 116 ASNS+/+ 癌细胞系。我们研究了 ASNS 缺失对肿瘤生长的影响,以及随后在雄性和雌性 Rag2/IL2RG 小鼠体内对代谢途径的重新布线:结果:ASNS缺失降低了雄性和雌性肿瘤小鼠的癌症负担(减少40%,q < 0.05),引发了包括葡萄糖生成在内的代谢重编程,但仅改善了雌性肿瘤小鼠的存活率(30天的中位存活率,q < 0.05)。转录组分析表明,在HCT116 ASNS-/-异种移植的雄性和雌性小鼠肿瘤中,G蛋白偶联雌激素受体(GPER1)上调。雌二醇在体外激活ASNS存在下的GPER1,并抑制肿瘤生长:我们的研究表明,高 ASNS 女性 CRC 患者的生存率较低可能是由于新陈代谢重编程导致了肿瘤的持续生长。这些发现具有转化意义,因为 ASNS/GPER1 信号转导可能是未来改善女性 CRC 患者生存率的治疗靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Growth characteristics of HCT116 xenografts lacking asparagine synthetase vary according to sex.

Background: Sex-related differences in colorectal (CRC) incidence and mortality are well-documented. However, the impact of sex on metabolic pathways that drive cancer growth is not well understood. High expression of asparagine synthetase (ASNS) is associated with inferior survival for female CRC patients only. Here, we used a CRISPR/Cas9 technology to generate HCT116 ASNS-/- and HCT 116 ASNS+/+ cancer cell lines. We examine the effects of ASNS deletion on tumor growth and the subsequent rewiring of metabolic pathways in male and female Rag2/IL2RG mice.

Results: ASNS loss reduces cancer burden in male and female tumor-bearing mice (40% reduction, q < 0.05), triggers metabolic reprogramming including gluconeogenesis, but confers a survival improvement (30 days median survival, q < 0.05) in female tumor-bearing mice alone. Transcriptomic analyses revealed upregulation of G-protein coupled estrogen receptor (GPER1) in tumors from male and female mice with HCT116 ASNS-/- xenograft. Estradiol activates GPER1 in vitro in the presence of ASNS and suppresses tumor growth.

Conclusions: Our study indicates that inferior survival for female CRC patients with high ASNS may be due to metabolic reprogramming that sustains tumor growth. These findings have translational relevance as ASNS/GPER1 signaling could be a future therapeutic target to improve the survival of female CRC patients.

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
Human Genomics
Human Genomics GENETICS & HEREDITY-
CiteScore
6.00
自引率
2.20%
发文量
55
审稿时长
11 weeks
期刊介绍: Human Genomics is a peer-reviewed, open access, online journal that focuses on the application of genomic analysis in all aspects of human health and disease, as well as genomic analysis of drug efficacy and safety, and comparative genomics. Topics covered by the journal include, but are not limited to: pharmacogenomics, genome-wide association studies, genome-wide sequencing, exome sequencing, next-generation deep-sequencing, functional genomics, epigenomics, translational genomics, expression profiling, proteomics, bioinformatics, animal models, statistical genetics, genetic epidemiology, human population genetics and comparative genomics.
期刊最新文献
Genetic history and biological adaptive landscape of the Tujia people inferred from shared haplotypes and alleles The cryptic complex rearrangements involving the DMD gene: etiologic clues about phenotypical differences revealed by optical genome mapping Whole-exome sequencing to identify causative variants in juvenile sudden cardiac death Integrated multiomics revealed adenosine signaling predict immunotherapy response and regulate tumor ecosystem of melanoma Reply to correspondence by Deora et al. in Human Genomics 18, article no.: 52 (2024): critical insights on “Association of the C allele of rs479200 in the EGLN1 gene with COVID‑19 severity in Indian population: a novel finding”
×
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