TRPM7激酶的失活靶向人类CML细胞中AKT信号传导和环氧合酶-2的表达。

IF 5.1 Q2 CELL BIOLOGY
Function (Oxford, England) Pub Date : 2023-09-15 eCollection Date: 2023-01-01 DOI:10.1093/function/zqad053
Birgit Hoeger, Wiebke Nadolni, Sarah Hampe, Kilian Hoelting, Marco Fraticelli, Nadja Zaborsky, Anna Madlmayr, Viktoria Sperrer, Laura Fraticelli, Lynda Addington, Dirk Steinritz, Vladimir Chubanov, Roland Geisberger, Richard Greil, Andreas Breit, Ingrid Boekhoff, Thomas Gudermann, Susanna Zierler
{"title":"TRPM7激酶的失活靶向人类CML细胞中AKT信号传导和环氧合酶-2的表达。","authors":"Birgit Hoeger,&nbsp;Wiebke Nadolni,&nbsp;Sarah Hampe,&nbsp;Kilian Hoelting,&nbsp;Marco Fraticelli,&nbsp;Nadja Zaborsky,&nbsp;Anna Madlmayr,&nbsp;Viktoria Sperrer,&nbsp;Laura Fraticelli,&nbsp;Lynda Addington,&nbsp;Dirk Steinritz,&nbsp;Vladimir Chubanov,&nbsp;Roland Geisberger,&nbsp;Richard Greil,&nbsp;Andreas Breit,&nbsp;Ingrid Boekhoff,&nbsp;Thomas Gudermann,&nbsp;Susanna Zierler","doi":"10.1093/function/zqad053","DOIUrl":null,"url":null,"abstract":"<p><p>Cyclooxygenase-2 (COX-2) is a key regulator of inflammation. High constitutive <i>COX-2</i> expression enhances survival and proliferation of cancer cells, and adversely impacts antitumor immunity. The expression of <i>COX-2</i> is modulated by various signaling pathways. Recently, we identified the melastatin-like transient-receptor-potential-7 (TRPM7) channel-kinase as modulator of immune homeostasis. TRPM7 protein is essential for leukocyte proliferation and differentiation, and upregulated in several cancers. It comprises of a cation channel and an atypical α-kinase, linked to inflammatory cell signals and associated with hallmarks of tumor progression. A role in leukemia has not been established, and signaling pathways are yet to be deciphered. We show that inhibiting TRPM7 channel-kinase in chronic myeloid leukemia (CML) cells results in reduced constitutive <i>COX-2</i> expression. By utilizing a CML-derived cell line, HAP1, harboring CRISPR/Cas9-mediated TRPM7 knockout, or a point mutation inactivating TRPM7 kinase, we could link this to reduced activation of AKT serine/threonine kinase and mothers against decapentaplegic homolog 2 (SMAD2). We identified AKT as a direct in vitro substrate of TRPM7 kinase. Pharmacologic blockade of TRPM7 in wildtype HAP1 cells confirmed the effect on <i>COX-2</i> via altered AKT signaling. Addition of an AKT activator on TRPM7 kinase-dead cells reconstituted the wildtype phenotype. Inhibition of TRPM7 resulted in reduced phosphorylation of AKT and diminished <i>COX-2</i> expression in peripheral blood mononuclear cells derived from CML patients, and reduced proliferation in patient-derived CD34<sup>+</sup> cells. These results highlight a role of TRPM7 kinase in AKT-driven <i>COX-2</i> expression and suggest a beneficial potential of TRPM7 blockade in COX-2-related inflammation and malignancy.</p>","PeriodicalId":73119,"journal":{"name":"Function (Oxford, England)","volume":null,"pages":null},"PeriodicalIF":5.1000,"publicationDate":"2023-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6d/e8/zqad053.PMC10541797.pdf","citationCount":"0","resultStr":"{\"title\":\"Inactivation of TRPM7 Kinase Targets AKT Signaling and Cyclooxygenase-2 Expression in Human CML Cells.\",\"authors\":\"Birgit Hoeger,&nbsp;Wiebke Nadolni,&nbsp;Sarah Hampe,&nbsp;Kilian Hoelting,&nbsp;Marco Fraticelli,&nbsp;Nadja Zaborsky,&nbsp;Anna Madlmayr,&nbsp;Viktoria Sperrer,&nbsp;Laura Fraticelli,&nbsp;Lynda Addington,&nbsp;Dirk Steinritz,&nbsp;Vladimir Chubanov,&nbsp;Roland Geisberger,&nbsp;Richard Greil,&nbsp;Andreas Breit,&nbsp;Ingrid Boekhoff,&nbsp;Thomas Gudermann,&nbsp;Susanna Zierler\",\"doi\":\"10.1093/function/zqad053\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Cyclooxygenase-2 (COX-2) is a key regulator of inflammation. High constitutive <i>COX-2</i> expression enhances survival and proliferation of cancer cells, and adversely impacts antitumor immunity. The expression of <i>COX-2</i> is modulated by various signaling pathways. Recently, we identified the melastatin-like transient-receptor-potential-7 (TRPM7) channel-kinase as modulator of immune homeostasis. TRPM7 protein is essential for leukocyte proliferation and differentiation, and upregulated in several cancers. It comprises of a cation channel and an atypical α-kinase, linked to inflammatory cell signals and associated with hallmarks of tumor progression. A role in leukemia has not been established, and signaling pathways are yet to be deciphered. We show that inhibiting TRPM7 channel-kinase in chronic myeloid leukemia (CML) cells results in reduced constitutive <i>COX-2</i> expression. By utilizing a CML-derived cell line, HAP1, harboring CRISPR/Cas9-mediated TRPM7 knockout, or a point mutation inactivating TRPM7 kinase, we could link this to reduced activation of AKT serine/threonine kinase and mothers against decapentaplegic homolog 2 (SMAD2). We identified AKT as a direct in vitro substrate of TRPM7 kinase. Pharmacologic blockade of TRPM7 in wildtype HAP1 cells confirmed the effect on <i>COX-2</i> via altered AKT signaling. Addition of an AKT activator on TRPM7 kinase-dead cells reconstituted the wildtype phenotype. Inhibition of TRPM7 resulted in reduced phosphorylation of AKT and diminished <i>COX-2</i> expression in peripheral blood mononuclear cells derived from CML patients, and reduced proliferation in patient-derived CD34<sup>+</sup> cells. These results highlight a role of TRPM7 kinase in AKT-driven <i>COX-2</i> expression and suggest a beneficial potential of TRPM7 blockade in COX-2-related inflammation and malignancy.</p>\",\"PeriodicalId\":73119,\"journal\":{\"name\":\"Function (Oxford, England)\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":5.1000,\"publicationDate\":\"2023-09-15\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6d/e8/zqad053.PMC10541797.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Function (Oxford, England)\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1093/function/zqad053\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2023/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q2\",\"JCRName\":\"CELL BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Function (Oxford, England)","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1093/function/zqad053","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/1/1 0:00:00","PubModel":"eCollection","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

环氧合酶-2(COX-2)是炎症的关键调节因子。高组成型COX-2表达增强癌症细胞的存活和增殖,并对抗肿瘤免疫产生不利影响。COX-2的表达受到多种信号通路的调节。最近,我们鉴定了美司他汀样瞬时受体电位7(TRPM7)通道激酶作为免疫稳态的调节剂。TRPM7蛋白是白细胞增殖和分化所必需的,并在几种癌症中上调。它由阳离子通道和非典型α-激酶组成,与炎症细胞信号有关,并与肿瘤进展的标志物有关。在白血病中的作用尚未确定,信号通路尚待破译。我们发现在慢性粒细胞白血病(CML)细胞中抑制TRPM7通道激酶导致组成型COX-2表达降低。通过利用携带CRISPR/Cas9介导的TRPM7敲除的CML衍生细胞系HAP1,或使TRPM7激酶失活的点突变,我们可以将其与AKT丝氨酸/苏氨酸激酶的活化减少和母亲对抗脑卒中同源物2(SMAD2)联系起来。我们确定AKT是TRPM7激酶的直接体外底物。野生型HAP1细胞中TRPM7的药理学阻断证实了通过改变AKT信号对COX-2的作用。在TRPM7激酶死亡细胞上添加AKT激活剂重建了野生型表型。TRPM7的抑制导致来源于CML患者的外周血单核细胞中AKT的磷酸化减少和COX-2的表达减少,以及来源于患者的CD34+细胞的增殖减少。这些结果强调了TRPM7激酶在AKT驱动的COX-2表达中的作用,并表明TRPM7阻断在COX-2相关炎症和恶性肿瘤中的有益潜力。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Inactivation of TRPM7 Kinase Targets AKT Signaling and Cyclooxygenase-2 Expression in Human CML Cells.

Inactivation of TRPM7 Kinase Targets AKT Signaling and Cyclooxygenase-2 Expression in Human CML Cells.

Inactivation of TRPM7 Kinase Targets AKT Signaling and Cyclooxygenase-2 Expression in Human CML Cells.

Inactivation of TRPM7 Kinase Targets AKT Signaling and Cyclooxygenase-2 Expression in Human CML Cells.

Cyclooxygenase-2 (COX-2) is a key regulator of inflammation. High constitutive COX-2 expression enhances survival and proliferation of cancer cells, and adversely impacts antitumor immunity. The expression of COX-2 is modulated by various signaling pathways. Recently, we identified the melastatin-like transient-receptor-potential-7 (TRPM7) channel-kinase as modulator of immune homeostasis. TRPM7 protein is essential for leukocyte proliferation and differentiation, and upregulated in several cancers. It comprises of a cation channel and an atypical α-kinase, linked to inflammatory cell signals and associated with hallmarks of tumor progression. A role in leukemia has not been established, and signaling pathways are yet to be deciphered. We show that inhibiting TRPM7 channel-kinase in chronic myeloid leukemia (CML) cells results in reduced constitutive COX-2 expression. By utilizing a CML-derived cell line, HAP1, harboring CRISPR/Cas9-mediated TRPM7 knockout, or a point mutation inactivating TRPM7 kinase, we could link this to reduced activation of AKT serine/threonine kinase and mothers against decapentaplegic homolog 2 (SMAD2). We identified AKT as a direct in vitro substrate of TRPM7 kinase. Pharmacologic blockade of TRPM7 in wildtype HAP1 cells confirmed the effect on COX-2 via altered AKT signaling. Addition of an AKT activator on TRPM7 kinase-dead cells reconstituted the wildtype phenotype. Inhibition of TRPM7 resulted in reduced phosphorylation of AKT and diminished COX-2 expression in peripheral blood mononuclear cells derived from CML patients, and reduced proliferation in patient-derived CD34+ cells. These results highlight a role of TRPM7 kinase in AKT-driven COX-2 expression and suggest a beneficial potential of TRPM7 blockade in COX-2-related inflammation and malignancy.

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
CiteScore
5.70
自引率
0.00%
发文量
0
审稿时长
3 weeks
×
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