Kindlin-2 Regulates the Oncogenic Activities of Integrins and TGF-β In Triple Negative Breast Cancer Progression and Metastasis

Neelum Aziz Yousafzai, Lamyae El Khalki, Wei Wang, Justin Szpendyk, K. Sossey-Alaoui
{"title":"Kindlin-2 Regulates the Oncogenic Activities of Integrins and TGF-β In Triple Negative Breast Cancer Progression and Metastasis","authors":"Neelum Aziz Yousafzai, Lamyae El Khalki, Wei Wang, Justin Szpendyk, K. Sossey-Alaoui","doi":"10.21203/rs.3.rs-3914650/v1","DOIUrl":null,"url":null,"abstract":"Abstract Background Kindlin-2, an adaptor protein, is dysregulated in various human cancers, including triple negative breast cancer (TNBC), where it drives tumor progression and metastasis by influencing several cancer hallmarks. One well-established role of Kindlin-2 involves the regulation of integrin signaling, achieved by directly binding to the cytoplasmic tail of the integrin β subunit. In this study, we present novel insights into Kindlin-2's involvement in stabilizing the β1-Integrin:TGF-β type 1 receptor (TβRI) complexes, acting as a physical bridge that links β1-Integrin to TβRI. The loss of Kindlin-2 results in the degradation of this protein complex, leading to the inhibition of downstream oncogenic pathways. Methods Our methodology encompassed a diverse range of in vitro assays, including CRISPR/Cas9 gene editing, cell migration, 3D tumorsphere formation and invasion, solid binding, co-immunoprecipitation, cell adhesion and spreading assays, as well as western blot and flow cytometry analyses, utilizing MDA-MB-231 and 4T1 TNBC cell lines. Additionally, preclinical in vivo mouse models of TNBC tumor progression and metastasis were employed to substantiate our findings. Results The investigation revealed that the direct interaction between Kindlin-2 and β1-Integrin is mediated through the C-terminal F3 domain of Kindlin-2, while the interaction between Kindlin-2 and TβRI is facilitated through the F2 domain of Kindlin-2. Disruption of this bridge, achieved via CRISPR/Cas9-mediated knockout of Kindlin-2, led to the degradation of β1-Integrin and TβRI, resulting in the inhibition of oncogenic pathways downstream of both proteins, subsequently hindering tumor growth and metastasis. Treatment of Kindlin-2-deficient cells with the proteasome inhibitor MG-132 restored the expression of both β1-Integrin and TβRI. Furthermore, the rescue of Kindlin-2 expression reinstated their oncogenic activities both in vitro and in vivo. Conclusions This study identifies a novel function of Kindlin-2 in stabilizing the β1-Integrin:TβR1 complexes and regulating their downstream oncogenic signaling. The translational implications of these findings are substantial, potentially unveiling new therapeutically targeted pathways crucial for the treatment of TNBC tumors.","PeriodicalId":21039,"journal":{"name":"Research Square","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Research Square","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.21203/rs.3.rs-3914650/v1","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Abstract Background Kindlin-2, an adaptor protein, is dysregulated in various human cancers, including triple negative breast cancer (TNBC), where it drives tumor progression and metastasis by influencing several cancer hallmarks. One well-established role of Kindlin-2 involves the regulation of integrin signaling, achieved by directly binding to the cytoplasmic tail of the integrin β subunit. In this study, we present novel insights into Kindlin-2's involvement in stabilizing the β1-Integrin:TGF-β type 1 receptor (TβRI) complexes, acting as a physical bridge that links β1-Integrin to TβRI. The loss of Kindlin-2 results in the degradation of this protein complex, leading to the inhibition of downstream oncogenic pathways. Methods Our methodology encompassed a diverse range of in vitro assays, including CRISPR/Cas9 gene editing, cell migration, 3D tumorsphere formation and invasion, solid binding, co-immunoprecipitation, cell adhesion and spreading assays, as well as western blot and flow cytometry analyses, utilizing MDA-MB-231 and 4T1 TNBC cell lines. Additionally, preclinical in vivo mouse models of TNBC tumor progression and metastasis were employed to substantiate our findings. Results The investigation revealed that the direct interaction between Kindlin-2 and β1-Integrin is mediated through the C-terminal F3 domain of Kindlin-2, while the interaction between Kindlin-2 and TβRI is facilitated through the F2 domain of Kindlin-2. Disruption of this bridge, achieved via CRISPR/Cas9-mediated knockout of Kindlin-2, led to the degradation of β1-Integrin and TβRI, resulting in the inhibition of oncogenic pathways downstream of both proteins, subsequently hindering tumor growth and metastasis. Treatment of Kindlin-2-deficient cells with the proteasome inhibitor MG-132 restored the expression of both β1-Integrin and TβRI. Furthermore, the rescue of Kindlin-2 expression reinstated their oncogenic activities both in vitro and in vivo. Conclusions This study identifies a novel function of Kindlin-2 in stabilizing the β1-Integrin:TβR1 complexes and regulating their downstream oncogenic signaling. The translational implications of these findings are substantial, potentially unveiling new therapeutically targeted pathways crucial for the treatment of TNBC tumors.
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
Kindlin-2 在三阴性乳腺癌进展和转移过程中调控整合素和 TGF-β 的致癌活性
摘要 背景 Kindlin-2是一种适配蛋白,在包括三阴性乳腺癌(TNBC)在内的多种人类癌症中出现失调。Kindlin-2 的一个公认作用是通过直接与整合素 β 亚基的胞质部结合来调节整合素信号转导。在这项研究中,我们对 Kindlin-2 参与稳定 β1-Integrin:TGF-β 1 型受体(TβRI)复合物,充当连接 β1-Integrin 和 TβRI 的物理桥梁提出了新的见解。Kindlin-2 的缺失会导致该蛋白复合物降解,从而抑制下游致癌途径。研究方法 我们的研究方法涵盖了多种体外检测,包括利用 MDA-MB-231 和 4T1 TNBC 细胞系进行 CRISPR/Cas9 基因编辑、细胞迁移、三维瘤球形成和侵袭、固体结合、共免疫沉淀、细胞粘附和扩散检测,以及 Western 印迹和流式细胞术分析。此外,还采用了 TNBC 肿瘤进展和转移的临床前体内小鼠模型来证实我们的研究结果。研究结果表明,Kindlin-2 和 β1-Integrin 之间的直接相互作用是通过 Kindlin-2 的 C 端 F3 结构域介导的,而 Kindlin-2 和 TβRI 之间的相互作用是通过 Kindlin-2 的 F2 结构域促进的。通过CRISPR/Cas9介导的Kindlin-2基因敲除实现了对这一桥梁的破坏,导致了β1-Integrin和TβRI的降解,从而抑制了这两种蛋白下游的致癌通路,继而阻碍了肿瘤的生长和转移。用蛋白酶体抑制剂MG-132处理Kindlin-2缺陷细胞可恢复β1-Integrin和TβRI的表达。此外,Kindlin-2 的表达得到挽救后,它们在体外和体内的致癌活性都得到了恢复。结论 本研究发现了 Kindlin-2 在稳定 β1-Integrin:TβR1 复合物和调节其下游致癌信号转导方面的新功能。这些发现的转化意义重大,有可能揭示治疗 TNBC 肿瘤的新靶向途径。
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 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Disparity in temporal and spatial relationships between resting-state electrophysiological and fMRI signals. Acute sympathetic activation blunts the hyperemic and vasodilatory response to passive leg movement Pharmacological PINK1 activation ameliorates Pathology in Parkinson’s Disease models Blink-related arousal network surges are shaped by cortical vigilance states Radiomics-Based Predictive Nomogram for Assessing the Risk of Intracranial Aneurysms
×
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