首页 > 最新文献

Cell Communication and Signaling最新文献

英文 中文
Menin-driven mTOR signaling sustains taxane resistance in CRPC and reveals a targetable vulnerability for combination therapy. menin驱动的mTOR信号传导维持CRPC中的紫杉烷耐药,并揭示了联合治疗的可靶向脆弱性。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-18 DOI: 10.1186/s12964-025-02594-1
Ipek Bulut, Buse Cevatemre, Neslihan Yuksel-Catal, Hamzah Syed, Tamer Onder, Ceyda Acilan

Prostate cancer (PC) progression is predominantly driven by androgen signaling, making androgen deprivation therapy (ADT) the standard treatment. However, the transition to castration-resistant prostate cancer (CRPC) significantly reduces ADT efficacy. While taxanes such as docetaxel (Dtx) and cabazitaxel (Cbz) are widely employed, therapeutic resistance remains a major clinical obstacle. To address this, we established docetaxel-resistant CRPC models and performed an epigenetic drug screen, identifying MLL-Menin and MLL-WDR5 inhibitors as potent agents capable of restoring taxane sensitivity through G2/M arrest and apoptosis induction. Functional depletion of Menin (MEN1) revealed its critical role in sustaining chemoresistance, selectively impairing proliferation in resistant cells, and preventing resistance emergence in parental lines.Integrative transcriptomic (RNA-seq) and epigenomic (CUT&RUN-seq) analyses suggested that Menin may play a regulatory role in mTOR signaling and E2F target pathways. Menin binding to the promoters of mTOR and Cyclin D1 was confirmed, and rescue experiments further validated its regulatory role. Analysis of TCGA datasets demonstrated co-expression of MEN1 and mTOR in advanced metastatic PC, supporting clinical relevance. Moreover, combination treatment with the mTOR inhibitor Torin-1 and docetaxel synergistically enhanced therapeutic response in Menin-depleted resistant cells. MEN1 knockdown also abrogated tumor growth in vivo.These findings identify Menin as one of the key mediator of taxane resistance in CRPC through the regulation of mTOR. Targeting Menin, alone or in combination with mTOR inhibition, represents a promising strategy to overcome resistance and improve therapeutic outcomes in taxane-refractory PC.

前列腺癌(PC)的进展主要由雄激素信号驱动,使雄激素剥夺疗法(ADT)成为标准治疗。然而,向去势抵抗性前列腺癌(CRPC)的转变会显著降低ADT的疗效。虽然紫杉醇类药物如多西他赛(Dtx)和卡巴他赛(Cbz)被广泛应用,但治疗耐药性仍然是一个主要的临床障碍。为了解决这个问题,我们建立了耐多西他赛的CRPC模型,并进行了表观遗传药物筛选,发现MLL-Menin和MLL-WDR5抑制剂是能够通过G2/M阻滞和诱导凋亡来恢复紫杉烷敏感性的有效药物。Menin (MEN1)的功能缺失揭示了其在维持化学耐药、选择性地损害耐药细胞增殖和防止亲本系产生耐药方面的关键作用。综合转录组学(RNA-seq)和表观基因组学(CUT&RUN-seq)分析表明,Menin可能在mTOR信号通路和E2F靶点通路中发挥调节作用。Menin与mTOR启动子和Cyclin D1的结合被证实,救援实验进一步验证了Menin的调控作用。TCGA数据集分析显示MEN1和mTOR在晚期转移性PC中共表达,支持临床相关性。此外,mTOR抑制剂Torin-1和多西他赛联合治疗可协同增强menin -贫耐药细胞的治疗反应。MEN1敲低也能抑制肿瘤在体内的生长。这些发现表明Menin是通过调控mTOR调控CRPC中紫杉烷抗性的关键媒介之一。靶向Menin,单独或联合mTOR抑制,是克服紫杉烷难治性PC耐药和改善治疗结果的有希望的策略。
{"title":"Menin-driven mTOR signaling sustains taxane resistance in CRPC and reveals a targetable vulnerability for combination therapy.","authors":"Ipek Bulut, Buse Cevatemre, Neslihan Yuksel-Catal, Hamzah Syed, Tamer Onder, Ceyda Acilan","doi":"10.1186/s12964-025-02594-1","DOIUrl":"https://doi.org/10.1186/s12964-025-02594-1","url":null,"abstract":"<p><p>Prostate cancer (PC) progression is predominantly driven by androgen signaling, making androgen deprivation therapy (ADT) the standard treatment. However, the transition to castration-resistant prostate cancer (CRPC) significantly reduces ADT efficacy. While taxanes such as docetaxel (Dtx) and cabazitaxel (Cbz) are widely employed, therapeutic resistance remains a major clinical obstacle. To address this, we established docetaxel-resistant CRPC models and performed an epigenetic drug screen, identifying MLL-Menin and MLL-WDR5 inhibitors as potent agents capable of restoring taxane sensitivity through G<sub>2</sub>/M arrest and apoptosis induction. Functional depletion of Menin (MEN1) revealed its critical role in sustaining chemoresistance, selectively impairing proliferation in resistant cells, and preventing resistance emergence in parental lines.Integrative transcriptomic (RNA-seq) and epigenomic (CUT&RUN-seq) analyses suggested that Menin may play a regulatory role in mTOR signaling and E2F target pathways. Menin binding to the promoters of mTOR and Cyclin D1 was confirmed, and rescue experiments further validated its regulatory role. Analysis of TCGA datasets demonstrated co-expression of MEN1 and mTOR in advanced metastatic PC, supporting clinical relevance. Moreover, combination treatment with the mTOR inhibitor Torin-1 and docetaxel synergistically enhanced therapeutic response in Menin-depleted resistant cells. MEN1 knockdown also abrogated tumor growth in vivo.These findings identify Menin as one of the key mediator of taxane resistance in CRPC through the regulation of mTOR. Targeting Menin, alone or in combination with mTOR inhibition, represents a promising strategy to overcome resistance and improve therapeutic outcomes in taxane-refractory PC.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145783643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nuclear β-catenin: molecular regulation, challenges and therapeutic opportunities. 核β-连环蛋白:分子调控、挑战和治疗机遇。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-18 DOI: 10.1186/s12964-025-02600-6
Beiyu Xie, Yue Cao, Wenxi Li, Ruihan Chen, Qiujing Zhou, Xin Xie

The spatiotemporal control of Wnt signaling orchestrates embryonic development and tissue homeostasis, while dysregulation of this pathway has been linked to a variety of diseases, including cancer, fibrosis and neurodegeneration. Although nuclear accumulation of β-catenin serves as a hallmark of Wnt pathway hyperactivation, the mechanisms controlling nuclear β-catenin remain obscure and are subjected to some conflicting hypotheses. In this narrative review, we summarize current understanding of the complex interplays fine-tunning the nucleocytoplasmic trafficking and subcellular distribution of β-catenin. We also present computational analysis to explore candidate molecules regulating nuclear β-catenin and suggest new perspectives to future study. Finally, we discuss how these insights could pave the way for mechanism-based approaches to target "undruggable" Wnt signaling for Wnt-driven diseases.

Wnt信号的时空调控协调了胚胎发育和组织稳态,而该通路的失调与多种疾病有关,包括癌症、纤维化和神经变性。尽管细胞核中β-catenin的积累是Wnt通路过度激活的标志,但控制核β-catenin的机制仍然不清楚,并且受到一些相互矛盾的假设的影响。在这篇叙述性的综述中,我们总结了目前对β-连环蛋白的核胞质转运和亚细胞分布的复杂相互作用的理解。我们还提出了计算分析来探索调节核β-连环蛋白的候选分子,并为未来的研究提出了新的观点。最后,我们讨论了这些见解如何为基于机制的方法铺平道路,以针对Wnt驱动疾病的“不可药物”Wnt信号。
{"title":"Nuclear β-catenin: molecular regulation, challenges and therapeutic opportunities.","authors":"Beiyu Xie, Yue Cao, Wenxi Li, Ruihan Chen, Qiujing Zhou, Xin Xie","doi":"10.1186/s12964-025-02600-6","DOIUrl":"https://doi.org/10.1186/s12964-025-02600-6","url":null,"abstract":"<p><p>The spatiotemporal control of Wnt signaling orchestrates embryonic development and tissue homeostasis, while dysregulation of this pathway has been linked to a variety of diseases, including cancer, fibrosis and neurodegeneration. Although nuclear accumulation of β-catenin serves as a hallmark of Wnt pathway hyperactivation, the mechanisms controlling nuclear β-catenin remain obscure and are subjected to some conflicting hypotheses. In this narrative review, we summarize current understanding of the complex interplays fine-tunning the nucleocytoplasmic trafficking and subcellular distribution of β-catenin. We also present computational analysis to explore candidate molecules regulating nuclear β-catenin and suggest new perspectives to future study. Finally, we discuss how these insights could pave the way for mechanism-based approaches to target \"undruggable\" Wnt signaling for Wnt-driven diseases.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145776580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fadraciclib, a CDK2/CDK9 inhibitor, shows efficacy in biliary tract cancer and synergistic potential with olaparib and JQ1 based on MCL1 expression. Fadraciclib是一种CDK2/CDK9抑制剂,基于MCL1的表达,显示出与奥拉帕尼和JQ1协同作用的胆道肿瘤疗效。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-17 DOI: 10.1186/s12964-025-02521-4
Jae-Min Kim, Ah-Rong Nam, Kyoung-Seok Oh, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Hyo Jung Kim, Su In Lee, Jeesun Yoon, Tae-Yong Kim, Do-Youn Oh

Background: Targeting CDKs has emerged as a significant strategy in cancer drug development. While CDK4/6 inhibitors have proven effective in several cancers, CDK2 and CDK9 inhibitors are under clinical trials. In biliary tract cancer (BTC), CDK2 and CDK9 expression levels are elevated compared to normal tissue. CDK9, a transcriptional CDK, regulates RNAPII, promoting the transcription of oncogenes, such as MCL1. Aberrant CDK activation contributes to cancer progression and apoptosis evasion in BTC. Notably, MCL1 is frequently amplified in intrahepatic cholangiocarcinoma (16-21%), supporting the therapeutic potential of CDK2 and CDK9. However, targeting CDK2/9 in BTC has not yet been explored. This study aimed to evaluate CDK2/9 inhibition and develop possible biomarker strategies in BTC.

Methods: Nine BTC cell lines (SNU245, SNU308, SNU478, SNU869, SNU1196, SNU2670, SNU2773, TFK1, and HUCCT1) were used. Fadraciclib (CDK2/9 inhibitor), olaparib (PARP inhibitor), and JQ1 (BRD4 inhibitor) were used. Anti-cancer effects were evaluated using MTT assay, colony formation assay, annexin-V assay, and cell cycle analysis. HR-mediated DNA damage repair was assessed using foci formation assay and DRGFP assay. Combination therapies were evaluated in vitro and in vivo.

Results: Fadraciclib was more effective in MCL1-High cells, reducing RNAPII phosphorylation and MCL1. Fadraciclib also inhibited HR gene transcription. Fadraciclib-olaparib combination showed synergy in MCL1-High cells and xenograft models. Conversely, in MCL1-Low cells, Fadraciclib upregulated BRD4, restoring RNAP II activity and oncogenes transcription. Combination of fadraciclib-JQ1 suppressed this restoration and showed synergy in vitro and in vivo.

Conclusions: MCL1-High BTCs are sensitive to CDK2/9 inhibition and benefit from combination with PARP inhibitor. In MCL1-Low BTCs, combining CDK2/9 inhibitor and BRD4 inhibitor may represent an optimal strategy for new drug development.

背景:靶向CDKs已成为癌症药物开发的重要策略。虽然CDK4/6抑制剂已被证明对几种癌症有效,但CDK2和CDK9抑制剂仍处于临床试验阶段。在胆道癌(BTC)中,CDK2和CDK9的表达水平与正常组织相比升高。CDK9是一种转录性CDK,可调控RNAPII,促进癌基因(如MCL1)的转录。异常的CDK激活有助于BTC的癌症进展和细胞凋亡逃避。值得注意的是,MCL1在肝内胆管癌中经常被扩增(16-21%),支持CDK2和CDK9的治疗潜力。然而,靶向CDK2/9在BTC中的作用尚未被探索。本研究旨在评估CDK2/9在BTC中的抑制作用,并开发可能的生物标志物策略。方法:采用9株BTC细胞系(SNU245、SNU308、SNU478、SNU869、SNU1196、SNU2670、SNU2773、TFK1、HUCCT1)。使用Fadraciclib (CDK2/9抑制剂)、olaparib (PARP抑制剂)和JQ1 (BRD4抑制剂)。采用MTT试验、菌落形成试验、膜联蛋白- v试验和细胞周期分析评估抗癌效果。采用病灶形成法和DRGFP法评估hr介导的DNA损伤修复。在体外和体内对联合疗法进行了评价。结果:Fadraciclib对MCL1- high细胞更有效,降低RNAPII磷酸化和MCL1。Fadraciclib还能抑制HR基因转录。Fadraciclib-olaparib联合在MCL1-High细胞和异种移植物模型中显示协同作用。相反,在MCL1-Low细胞中,Fadraciclib上调BRD4,恢复RNAP II活性和癌基因转录。fadraciclib-JQ1联合使用抑制了这种恢复,并在体外和体内表现出协同作用。结论:mcl1 -高btc对CDK2/9抑制敏感,与PARP抑制剂联合使用可获益。在MCL1-Low btc中,联合CDK2/9抑制剂和BRD4抑制剂可能是新药开发的最佳策略。
{"title":"Fadraciclib, a CDK2/CDK9 inhibitor, shows efficacy in biliary tract cancer and synergistic potential with olaparib and JQ1 based on MCL1 expression.","authors":"Jae-Min Kim, Ah-Rong Nam, Kyoung-Seok Oh, Ju-Hee Bang, Yoojin Jeong, Sea Young Choo, Hyo Jung Kim, Su In Lee, Jeesun Yoon, Tae-Yong Kim, Do-Youn Oh","doi":"10.1186/s12964-025-02521-4","DOIUrl":"10.1186/s12964-025-02521-4","url":null,"abstract":"<p><strong>Background: </strong>Targeting CDKs has emerged as a significant strategy in cancer drug development. While CDK4/6 inhibitors have proven effective in several cancers, CDK2 and CDK9 inhibitors are under clinical trials. In biliary tract cancer (BTC), CDK2 and CDK9 expression levels are elevated compared to normal tissue. CDK9, a transcriptional CDK, regulates RNAPII, promoting the transcription of oncogenes, such as MCL1. Aberrant CDK activation contributes to cancer progression and apoptosis evasion in BTC. Notably, MCL1 is frequently amplified in intrahepatic cholangiocarcinoma (16-21%), supporting the therapeutic potential of CDK2 and CDK9. However, targeting CDK2/9 in BTC has not yet been explored. This study aimed to evaluate CDK2/9 inhibition and develop possible biomarker strategies in BTC.</p><p><strong>Methods: </strong>Nine BTC cell lines (SNU245, SNU308, SNU478, SNU869, SNU1196, SNU2670, SNU2773, TFK1, and HUCCT1) were used. Fadraciclib (CDK2/9 inhibitor), olaparib (PARP inhibitor), and JQ1 (BRD4 inhibitor) were used. Anti-cancer effects were evaluated using MTT assay, colony formation assay, annexin-V assay, and cell cycle analysis. HR-mediated DNA damage repair was assessed using foci formation assay and DRGFP assay. Combination therapies were evaluated in vitro and in vivo.</p><p><strong>Results: </strong>Fadraciclib was more effective in MCL1-High cells, reducing RNAPII phosphorylation and MCL1. Fadraciclib also inhibited HR gene transcription. Fadraciclib-olaparib combination showed synergy in MCL1-High cells and xenograft models. Conversely, in MCL1-Low cells, Fadraciclib upregulated BRD4, restoring RNAP II activity and oncogenes transcription. Combination of fadraciclib-JQ1 suppressed this restoration and showed synergy in vitro and in vivo.</p><p><strong>Conclusions: </strong>MCL1-High BTCs are sensitive to CDK2/9 inhibition and benefit from combination with PARP inhibitor. In MCL1-Low BTCs, combining CDK2/9 inhibitor and BRD4 inhibitor may represent an optimal strategy for new drug development.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"530"},"PeriodicalIF":8.2,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12709703/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145776578","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CHI3L1 promotes cell division through activation of RhoA in lung cancer cells. 在肺癌细胞中,CHI3L1通过激活RhoA促进细胞分裂。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-17 DOI: 10.1186/s12964-025-02601-5
Ji Eun Yu, Jung-Hyun Shim, In Jun Yeo, Dong Ju Son, Jaesuk Yun, Key-Hwan Lim, Hanseul Park, Yoon-Seok Roh, Sang-Bae Han, Jin Tae Hong
{"title":"CHI3L1 promotes cell division through activation of RhoA in lung cancer cells.","authors":"Ji Eun Yu, Jung-Hyun Shim, In Jun Yeo, Dong Ju Son, Jaesuk Yun, Key-Hwan Lim, Hanseul Park, Yoon-Seok Roh, Sang-Bae Han, Jin Tae Hong","doi":"10.1186/s12964-025-02601-5","DOIUrl":"https://doi.org/10.1186/s12964-025-02601-5","url":null,"abstract":"","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145776651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GALNT3 is a novel target driving lymphomagenesis via O-glycosylation of FGFR2. GALNT3是通过FGFR2的o -糖基化驱动淋巴瘤发生的新靶点。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-17 DOI: 10.1186/s12964-025-02599-w
Ruixin Sun, Wenhan Wang, Yixuan Gao, Kasimujiang Aximujiang, Yan Li, Agida Innocent, Zizhen Xu, Jiao Ma

Despite the dual roles of GALNT3 in various cellular processes in tumorigenesis of solid tumors, the clinical and biological significance of GALNT3 in lymphomagenesis remains largely unknown. Herein, our bioinformatics analysis uncovers that GALNT3 dependent glucose metabolism was significantly elevated in DLBCL. Our DLBCL cohort study revealed that GALNT3 positivity is significantly associated with worse prognosis and clinical outcomes in DLBCL pathogenesis. Biologically, GALNT3 overexpression were found to promote DLBCL cell proliferation and cell cycle progression, while silencing GALNT3 inhibited lymphomagenesis. RNA-seq uncovers that GALNT3 upregulated FGFR2-MAPK signaling pathway in DLBCL. Moreover, protein mass spectrometry identified two potential O-Glycosylation sites of FGFR2 (Thr319 and Ser299). Further point mutation of glycosylation sites, confirmed that GALNT3 O-Glycosylates FGFR2 through Thr319. Besides, in vitro and in vivo rescue experiments demonstrated that Thr319 is indispensable for GALNT3-FGFR2-MAPK induced lymphomagenesis. In vitro pharmacological inhibition of FGFR2 with a selective inhibitor Futibatinib further demonstrated that it inhibited DLBCL cell growth, cell proliferation, induced cell cycle arrest, promoted cell apoptosis. Further in vivo study found that combination of Futibatinib with chemotherapy displayed better anti-tumor activity relative to single drug therapy in DLBCL treatment. Collectively, our data highlight the importance of considering the GALNT3-FGFR2-MAPK signaling axis as an attractive therapeutic target for lymphomagenesis. Besides, in vitro and in vivo rescue experiments demonstrated that Thr319 is indispensable for GALNT3-FGFR2-MAPK induced lymphomagenesis. In vitro pharmacological inhibition of FGFR2 with a selective inhibitor Futibatinib further demonstrated that it inhibited DLBCL cell growth, cell proliferation, induced cell cycle arrest, promoted cell apoptosis. Further in vivo study found that combination of Futibatinib with chemotherapy displayed better anti-tumor activity relative to single drug therapy in DLBCL treatment.Collectively, our data highlight the importance of considering the GALNT3-FGFR2-MAPK signaling axis as an attractive therapeutic target for lymphomagenesis.

尽管GALNT3在实体瘤发生的各种细胞过程中具有双重作用,但GALNT3在淋巴瘤发生中的临床和生物学意义仍然很大程度上未知。在此,我们的生物信息学分析发现,GALNT3依赖性葡萄糖代谢在DLBCL中显著升高。我们的DLBCL队列研究显示GALNT3阳性与DLBCL发病机制中较差的预后和临床结果显著相关。生物学上,GALNT3过表达可促进DLBCL细胞增殖和细胞周期进展,而沉默GALNT3可抑制淋巴瘤发生。RNA-seq发现GALNT3上调DLBCL中FGFR2-MAPK信号通路。此外,蛋白质谱鉴定出FGFR2的两个潜在o -糖基化位点(Thr319和Ser299)。糖基化位点进一步点突变,证实GALNT3 O-Glycosylates FGFR2通过Thr319。此外,体外和体内救援实验表明,Thr319对于GALNT3-FGFR2-MAPK诱导的淋巴瘤发生是必不可少的。选择性抑制剂Futibatinib体外药理抑制FGFR2进一步证明其抑制DLBCL细胞生长,细胞增殖,诱导细胞周期阻滞,促进细胞凋亡。进一步的体内研究发现,在DLBCL治疗中,富替替尼联合化疗比单药治疗具有更好的抗肿瘤活性。总的来说,我们的数据强调了将GALNT3-FGFR2-MAPK信号轴作为淋巴瘤发生的有吸引力的治疗靶点的重要性。此外,体外和体内救援实验表明,Thr319对于GALNT3-FGFR2-MAPK诱导的淋巴瘤发生是必不可少的。选择性抑制剂Futibatinib体外药理抑制FGFR2进一步证明其抑制DLBCL细胞生长,细胞增殖,诱导细胞周期阻滞,促进细胞凋亡。进一步的体内研究发现,在DLBCL治疗中,富替替尼联合化疗比单药治疗具有更好的抗肿瘤活性。总的来说,我们的数据强调了将GALNT3-FGFR2-MAPK信号轴作为淋巴瘤发生的有吸引力的治疗靶点的重要性。
{"title":"GALNT3 is a novel target driving lymphomagenesis via O-glycosylation of FGFR2.","authors":"Ruixin Sun, Wenhan Wang, Yixuan Gao, Kasimujiang Aximujiang, Yan Li, Agida Innocent, Zizhen Xu, Jiao Ma","doi":"10.1186/s12964-025-02599-w","DOIUrl":"https://doi.org/10.1186/s12964-025-02599-w","url":null,"abstract":"<p><p>Despite the dual roles of GALNT3 in various cellular processes in tumorigenesis of solid tumors, the clinical and biological significance of GALNT3 in lymphomagenesis remains largely unknown. Herein, our bioinformatics analysis uncovers that GALNT3 dependent glucose metabolism was significantly elevated in DLBCL. Our DLBCL cohort study revealed that GALNT3 positivity is significantly associated with worse prognosis and clinical outcomes in DLBCL pathogenesis. Biologically, GALNT3 overexpression were found to promote DLBCL cell proliferation and cell cycle progression, while silencing GALNT3 inhibited lymphomagenesis. RNA-seq uncovers that GALNT3 upregulated FGFR2-MAPK signaling pathway in DLBCL. Moreover, protein mass spectrometry identified two potential O-Glycosylation sites of FGFR2 (Thr319 and Ser299). Further point mutation of glycosylation sites, confirmed that GALNT3 O-Glycosylates FGFR2 through Thr319. Besides, in vitro and in vivo rescue experiments demonstrated that Thr319 is indispensable for GALNT3-FGFR2-MAPK induced lymphomagenesis. In vitro pharmacological inhibition of FGFR2 with a selective inhibitor Futibatinib further demonstrated that it inhibited DLBCL cell growth, cell proliferation, induced cell cycle arrest, promoted cell apoptosis. Further in vivo study found that combination of Futibatinib with chemotherapy displayed better anti-tumor activity relative to single drug therapy in DLBCL treatment. Collectively, our data highlight the importance of considering the GALNT3-FGFR2-MAPK signaling axis as an attractive therapeutic target for lymphomagenesis. Besides, in vitro and in vivo rescue experiments demonstrated that Thr319 is indispensable for GALNT3-FGFR2-MAPK induced lymphomagenesis. In vitro pharmacological inhibition of FGFR2 with a selective inhibitor Futibatinib further demonstrated that it inhibited DLBCL cell growth, cell proliferation, induced cell cycle arrest, promoted cell apoptosis. Further in vivo study found that combination of Futibatinib with chemotherapy displayed better anti-tumor activity relative to single drug therapy in DLBCL treatment.Collectively, our data highlight the importance of considering the GALNT3-FGFR2-MAPK signaling axis as an attractive therapeutic target for lymphomagenesis.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145776602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A proteogenomic gene signature defines prognostic subgroups highlighting PI3K/AKT/mTOR signaling pathway as a therapeutic vulnerability in myeloid malignancies. 一个蛋白质基因组基因标记定义了预后亚组,突出PI3K/AKT/mTOR信号通路作为髓系恶性肿瘤的治疗易感性。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-16 DOI: 10.1186/s12964-025-02568-3
Fan He, Shuyang Lin, Bei Gao, Varun Ramesh, Alexander B Kim, Tim Kong, Daniel A C Fisher, Christopher T Letson, Molly Brakhane, Mary Fulbright, Yanbo Yu, Marco Sardiello, Jorge Di Paola, Stephen M Sykes, Stephen T Oh

Introduction: Myeloid malignancies, including acute myeloid leukemia (AML) and myeloproliferative neoplasms (MPN), exhibit overlapping pathophysiology. Chronic MPNs can transform into secondary AML (sAML), which is associated with poor prognosis and limited treatment options. However, the process and prognostic significance of leukemic transformation remain incompletely understood.

Method: Through a two-sample bidirectional Mendelian randomization (MR) analysis, we showed that genetic liability to MPN significantly predicts the risk of developing AML, establishing MPN as the precursor to leukemia. To identify mediators of this risk, we integrated population-level plasma proteomics data, identifying 55 proteins associated with MPN. Upon integrative analysis with the BEAT-AML cohort, we developed a prognostic proteogenomic gene signature, showing that higher expression of CDCP1, CRISP3, and DXCR, alongside lower MPO levels, correlates with worse AML outcomes. We further performed pharmacogenomic analysis to identify vulnerability to PI3K/AKT/mTOR signaling pathway inhibition in high-risk AML. In vitro and in vivo experiments validated the efficacy of mTOR inhibition in myeloid malignancies.

Results: This gene signature effectively stratified patients by risk, with significant survival differences across the BEAT-AML and TCGA-LAML cohorts, and revealed immune alterations in high-risk groups, including elevated monocyte prevalence and cytokine signaling activity. Single-cell RNA sequencing (scRNA-seq) further suggested enrichment of these genes in progenitor cells and AML blasts. Drug sensitivity predictions suggested that high-risk AML patients may be particularly responsive to PI3K/AKT/mTOR signaling pathway inhibitors. Consistently, we observed upregulation of the genes in cell line models harboring MPN and AML mutations, which was suppressible via dual PI3K/mTOR inhibitor Omipalisib. The efficiency of PI3K/mTOR inhibition in myeloid malignancies was further corroborated by results from multiple in vivo models.

Conclusion: Together, our findings revealed shared molecular features across MPN and AML, identified a prognostic gene signature for risk stratification, and provided rationale for PI3K/mTOR inhibition as a promising therapeutic strategy in myeloid malignancies.

髓系恶性肿瘤,包括急性髓系白血病(AML)和骨髓增生性肿瘤(MPN),表现出重叠的病理生理。慢性mpn可转化为继发性AML (sAML),这与预后不良和治疗选择有限有关。然而,白血病转化的过程和预后意义仍不完全清楚。方法:通过双样本双向孟德尔随机化(MR)分析,我们发现MPN的遗传倾向性可以显著预测发生AML的风险,从而确定MPN是白血病的前体。为了确定这种风险的介质,我们整合了人群水平的血浆蛋白质组学数据,确定了55种与MPN相关的蛋白质。通过与BEAT-AML队列的综合分析,我们开发了预后蛋白质基因组基因标记,显示CDCP1、CRISP3和DXCR的高表达以及较低的MPO水平与较差的AML结果相关。我们进一步进行了药物基因组学分析,以确定高风险AML对PI3K/AKT/mTOR信号通路抑制的易感性。体外和体内实验验证了mTOR抑制髓系恶性肿瘤的有效性。结果:该基因标记有效地将患者按风险分层,在BEAT-AML和TCGA-LAML队列中存在显着的生存差异,并揭示了高危组的免疫改变,包括单核细胞患病率升高和细胞因子信号传导活性升高。单细胞RNA测序(scRNA-seq)进一步表明这些基因在祖细胞和AML母细胞中富集。药物敏感性预测表明,高风险AML患者可能对PI3K/AKT/mTOR信号通路抑制剂特别敏感。我们一致地观察到,在MPN和AML突变的细胞系模型中,基因上调,这是通过双PI3K/mTOR抑制剂Omipalisib抑制的。多个体内模型的结果进一步证实了PI3K/mTOR抑制髓系恶性肿瘤的有效性。结论:总之,我们的研究结果揭示了MPN和AML的共同分子特征,确定了风险分层的预后基因特征,并为PI3K/mTOR抑制作为髓系恶性肿瘤的一种有希望的治疗策略提供了理论依据。
{"title":"A proteogenomic gene signature defines prognostic subgroups highlighting PI3K/AKT/mTOR signaling pathway as a therapeutic vulnerability in myeloid malignancies.","authors":"Fan He, Shuyang Lin, Bei Gao, Varun Ramesh, Alexander B Kim, Tim Kong, Daniel A C Fisher, Christopher T Letson, Molly Brakhane, Mary Fulbright, Yanbo Yu, Marco Sardiello, Jorge Di Paola, Stephen M Sykes, Stephen T Oh","doi":"10.1186/s12964-025-02568-3","DOIUrl":"https://doi.org/10.1186/s12964-025-02568-3","url":null,"abstract":"<p><strong>Introduction: </strong>Myeloid malignancies, including acute myeloid leukemia (AML) and myeloproliferative neoplasms (MPN), exhibit overlapping pathophysiology. Chronic MPNs can transform into secondary AML (sAML), which is associated with poor prognosis and limited treatment options. However, the process and prognostic significance of leukemic transformation remain incompletely understood.</p><p><strong>Method: </strong>Through a two-sample bidirectional Mendelian randomization (MR) analysis, we showed that genetic liability to MPN significantly predicts the risk of developing AML, establishing MPN as the precursor to leukemia. To identify mediators of this risk, we integrated population-level plasma proteomics data, identifying 55 proteins associated with MPN. Upon integrative analysis with the BEAT-AML cohort, we developed a prognostic proteogenomic gene signature, showing that higher expression of CDCP1, CRISP3, and DXCR, alongside lower MPO levels, correlates with worse AML outcomes. We further performed pharmacogenomic analysis to identify vulnerability to PI3K/AKT/mTOR signaling pathway inhibition in high-risk AML. In vitro and in vivo experiments validated the efficacy of mTOR inhibition in myeloid malignancies.</p><p><strong>Results: </strong>This gene signature effectively stratified patients by risk, with significant survival differences across the BEAT-AML and TCGA-LAML cohorts, and revealed immune alterations in high-risk groups, including elevated monocyte prevalence and cytokine signaling activity. Single-cell RNA sequencing (scRNA-seq) further suggested enrichment of these genes in progenitor cells and AML blasts. Drug sensitivity predictions suggested that high-risk AML patients may be particularly responsive to PI3K/AKT/mTOR signaling pathway inhibitors. Consistently, we observed upregulation of the genes in cell line models harboring MPN and AML mutations, which was suppressible via dual PI3K/mTOR inhibitor Omipalisib. The efficiency of PI3K/mTOR inhibition in myeloid malignancies was further corroborated by results from multiple in vivo models.</p><p><strong>Conclusion: </strong>Together, our findings revealed shared molecular features across MPN and AML, identified a prognostic gene signature for risk stratification, and provided rationale for PI3K/mTOR inhibition as a promising therapeutic strategy in myeloid malignancies.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145770075","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparison of human cerebral organoids infected with wild-type Zika versus attenuated DN-2 virus strains uncovers differences in host immune responses. 野生型寨卡病毒与减毒DN-2病毒株感染的人脑类器官的比较揭示了宿主免疫反应的差异。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-16 DOI: 10.1186/s12964-025-02588-z
Yun Ying Choo, Tuan Zea Tan, Summer Lixin Zhang, Hwee Cheng Tan, Eng Eong Ooi, John Jia En Chua

Zika virus (ZIKV), a neurotropic virus, poses significant global health challenges because of its ability to cause severe neurological complications and congenital brain abnormalities. Prenatal ZIKV infection impairs early brain development. However, details of the molecular mechanisms driving their virulence remain incompletely understood. Here, we performed comparatively analyses of infection outcomes caused by 2 different ZIKV strains - wild-type (WT) ZIKV versus an attenuated strain (DN-2) - in human cerebral organoids (hCOs) to dissect host responses that could contribute to pathogenicity. Although both viral strains productively infected hCOs with indiscernible gross pathological changes, differences in host responses and subtypes of cells infected discriminate WT ZIKV from DN-2 infections. Single cell RNA sequencing analyses uncovered differently expressed genes (DEGs) that were common to both virus strains, as well as DEGs that were specific to either WT ZIKV or DN-2. For common DEGs, WT ZIKV infections elicited stronger expression of key genes involved in host immune response pathways, such as IFIT2, DDX60, OAS1 and XAF1. Moreover, analyses of WT ZIKV-specific upregulated DEGs uncovered mobilisation of additional innate immune response pathways not found in DN-2. Additionally, while WT ZIKV infected a broad range of cell types in hCOs, DN-2 infections were predominantly limited to radial glial, which are neuroprogenitors essential for neurogenesis. These results highlight that differences in host immune responses and tropism distinguish neurotropic WT ZIKV and attenuated DN-2 infections, underscoring the mechanistic differences involved in pathogenicity. Our study provides further insights into mechanisms used by neurotrophic ZIKV to drive neuropathogenesis during infection of the developing brain.

寨卡病毒(ZIKV)是一种嗜神经病毒,由于能够引起严重的神经系统并发症和先天性脑异常,对全球卫生构成重大挑战。产前寨卡病毒感染会损害早期大脑发育。然而,驱动其毒力的分子机制的细节仍然不完全清楚。在这里,我们对2种不同的ZIKV菌株——野生型(WT)与减毒株(DN-2)——在人脑类器官(hCOs)中引起的感染结果进行了比较分析,以剖析可能导致致病性的宿主反应。尽管这两种病毒株都能有效感染hCOs,但宿主反应和感染细胞亚型的差异将WT ZIKV与DN-2感染区分开来。单细胞RNA测序分析揭示了两种病毒株共有的不同表达基因(DEGs),以及WT ZIKV或DN-2特有的DEGs。对于常见的DEGs, WT ZIKV感染引发了参与宿主免疫反应途径的关键基因,如IFIT2、DDX60、OAS1和XAF1的更强表达。此外,对WT zikv特异性上调的DEGs的分析揭示了DN-2中未发现的额外先天免疫反应途径的动员。此外,虽然WT ZIKV在hCOs中感染了广泛的细胞类型,但DN-2感染主要局限于放射状胶质细胞,这是神经发生所必需的神经祖细胞。这些结果强调了宿主免疫反应和嗜神经型WT ZIKV和减毒DN-2感染的差异,强调了与致病性有关的机制差异。我们的研究为神经营养性ZIKV在发育中的大脑感染过程中驱动神经发病机制提供了进一步的见解。
{"title":"Comparison of human cerebral organoids infected with wild-type Zika versus attenuated DN-2 virus strains uncovers differences in host immune responses.","authors":"Yun Ying Choo, Tuan Zea Tan, Summer Lixin Zhang, Hwee Cheng Tan, Eng Eong Ooi, John Jia En Chua","doi":"10.1186/s12964-025-02588-z","DOIUrl":"https://doi.org/10.1186/s12964-025-02588-z","url":null,"abstract":"<p><p>Zika virus (ZIKV), a neurotropic virus, poses significant global health challenges because of its ability to cause severe neurological complications and congenital brain abnormalities. Prenatal ZIKV infection impairs early brain development. However, details of the molecular mechanisms driving their virulence remain incompletely understood. Here, we performed comparatively analyses of infection outcomes caused by 2 different ZIKV strains - wild-type (WT) ZIKV versus an attenuated strain (DN-2) - in human cerebral organoids (hCOs) to dissect host responses that could contribute to pathogenicity. Although both viral strains productively infected hCOs with indiscernible gross pathological changes, differences in host responses and subtypes of cells infected discriminate WT ZIKV from DN-2 infections. Single cell RNA sequencing analyses uncovered differently expressed genes (DEGs) that were common to both virus strains, as well as DEGs that were specific to either WT ZIKV or DN-2. For common DEGs, WT ZIKV infections elicited stronger expression of key genes involved in host immune response pathways, such as IFIT2, DDX60, OAS1 and XAF1. Moreover, analyses of WT ZIKV-specific upregulated DEGs uncovered mobilisation of additional innate immune response pathways not found in DN-2. Additionally, while WT ZIKV infected a broad range of cell types in hCOs, DN-2 infections were predominantly limited to radial glial, which are neuroprogenitors essential for neurogenesis. These results highlight that differences in host immune responses and tropism distinguish neurotropic WT ZIKV and attenuated DN-2 infections, underscoring the mechanistic differences involved in pathogenicity. Our study provides further insights into mechanisms used by neurotrophic ZIKV to drive neuropathogenesis during infection of the developing brain.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145764497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Temporal transcriptomic profiling reveals distinct age-associated gene expression signatures in gonads under reduced insulin/IGF-1 signaling in Caenorhabditis elegans. 在秀丽隐杆线虫中,时间转录组分析揭示了胰岛素/IGF-1信号减少的性腺中明显的年龄相关基因表达特征。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-15 DOI: 10.1186/s12964-025-02510-7
Neha Kaushik, Soumya Rastogi, Shivani Kapadia, Vaibhav Jain, Sonia Verma, Deepak Pandey, Mona Sharma, Surabhi Gupta, Ashutosh Halder, Simon Gregory, Neeraj Kumar

Background: Age-related decline in reproductive function is a hallmark of organismal aging, yet the molecular mechanisms driving this process remain incompletely understood. The insulin/IGF-1 signaling (IIS) pathway is highly conserved and influences both lifespan and reproductive aging in Caenorhabditis elegans, where reduced IIS extends reproductive span. While prior studies have examined isolated tissues or time points, a comprehensive temporal analysis of gonadal transcriptional dynamics under reduced IIS has been lacking. Here, we compared IIS-dependent regulation of the gonadal transcriptome with that of other somatic tissues to uncover tissue-specific mechanisms of reproductive aging.

Methods: Bulk RNA sequencing was performed on distal gonads dissected above the spermatheca from wild-type N2 and daf-2(e1370) mutant animals, a well-established model of reduced IIS. Samples were collected at four physiologically relevant adult stages-Day 1 (young adult), Day 2, Day 6, and Day 10-covering early to late reproductive periods. In parallel, whole-worm RNA-seq was conducted for N2 and daf-2 at Day 1 and Day 10 to enable systemic comparisons. Differential gene expression analyses identified IIS-responsive transcripts that were either gonad-specific or non-gonadal. Expression datasets were further analyzed using self-organizing maps (SOMs) with hierarchical clustering. Gene network construction, functional enrichment, transcription factor enrichment, and conservation analyses were performed, and differential expression profiles were integrated with publicly available germline-, gamete-, and somatic tissue-enriched datasets.

Results: Temporal transcriptomic profiling revealed distinct IIS-dependent expression trajectories in gonadal versus non-gonadal datasets. SOM-based clustering resolved temporally regulated expression modules, while network and enrichment analyses uncovered a multilayered regulatory architecture within the gonad. Gonadal expression was enriched for structural, extracellular matrix, and signaling pathway genes, whereas non-gonadal data showed enrichment for stress response and longevity-associated pathways. Integration with germline-, gamete-, and somatic tissue-enriched datasets distinguished tissue-specific regulatory signatures. Importantly, IIS-regulated gonadal components included genes highly conserved with human orthologs.

Conclusion: This study provides a high-resolution temporal map of the gonadal transcriptome under reduced IIS and highlights gene modules potentially critical for reproductive maintenance. These findings offer a resource for dissecting tissue-specific aging programs and insulin-dependent regulation of reproductive health.

背景:与年龄相关的生殖功能下降是机体衰老的一个标志,但驱动这一过程的分子机制仍不完全清楚。胰岛素/IGF-1信号通路(IIS)是高度保守的,影响秀丽隐杆线虫的寿命和生殖衰老,其中IIS的减少延长了生殖期。虽然先前的研究已经检查了孤立的组织或时间点,但缺乏对IIS降低下性腺转录动力学的全面时间分析。在这里,我们比较了iis依赖性的性腺转录组调控与其他体细胞组织的调控,以揭示生殖衰老的组织特异性机制。方法:对野生型N2和daf-2(e1370)突变动物的精囊上方解剖的远端性腺进行大量RNA测序,daf-2(e1370)是一种建立的IIS减少模型。在4个生理上相关的成虫阶段——第1天(青年成虫)、第2天、第6天和第10天采集样本,覆盖生殖早期到晚期。同时,在第1天和第10天对N2和daf-2进行全虫rna测序,以便进行系统比较。差异基因表达分析确定了iis应答转录本,这些转录本要么是性腺特异性的,要么是非性腺特异性的。表达数据集进一步分析使用自组织地图(SOMs)与层次聚类。进行了基因网络构建、功能富集、转录因子富集和保守分析,并将差异表达谱与公开可用的种系、配子和体细胞组织富集数据集集成。结果:时间转录组分析揭示了在性腺和非性腺数据集中不同的iis依赖性表达轨迹。基于som的聚类解析了暂时调控的表达模块,而网络和富集分析揭示了性腺内的多层调控结构。性腺结构、细胞外基质和信号通路基因的表达丰富,而非性腺数据显示应激反应和长寿相关通路的表达丰富。整合生殖系、配子和体细胞组织丰富的数据集,区分组织特异性调控信号。重要的是,iis调节的性腺成分包括与人类同源物高度保守的基因。结论:本研究提供了IIS降低下性腺转录组的高分辨率时间图谱,并突出了可能对生殖维持至关重要的基因模块。这些发现为解剖组织特异性衰老程序和生殖健康的胰岛素依赖调节提供了资源。
{"title":"Temporal transcriptomic profiling reveals distinct age-associated gene expression signatures in gonads under reduced insulin/IGF-1 signaling in Caenorhabditis elegans.","authors":"Neha Kaushik, Soumya Rastogi, Shivani Kapadia, Vaibhav Jain, Sonia Verma, Deepak Pandey, Mona Sharma, Surabhi Gupta, Ashutosh Halder, Simon Gregory, Neeraj Kumar","doi":"10.1186/s12964-025-02510-7","DOIUrl":"10.1186/s12964-025-02510-7","url":null,"abstract":"<p><strong>Background: </strong>Age-related decline in reproductive function is a hallmark of organismal aging, yet the molecular mechanisms driving this process remain incompletely understood. The insulin/IGF-1 signaling (IIS) pathway is highly conserved and influences both lifespan and reproductive aging in Caenorhabditis elegans, where reduced IIS extends reproductive span. While prior studies have examined isolated tissues or time points, a comprehensive temporal analysis of gonadal transcriptional dynamics under reduced IIS has been lacking. Here, we compared IIS-dependent regulation of the gonadal transcriptome with that of other somatic tissues to uncover tissue-specific mechanisms of reproductive aging.</p><p><strong>Methods: </strong>Bulk RNA sequencing was performed on distal gonads dissected above the spermatheca from wild-type N2 and daf-2(e1370) mutant animals, a well-established model of reduced IIS. Samples were collected at four physiologically relevant adult stages-Day 1 (young adult), Day 2, Day 6, and Day 10-covering early to late reproductive periods. In parallel, whole-worm RNA-seq was conducted for N2 and daf-2 at Day 1 and Day 10 to enable systemic comparisons. Differential gene expression analyses identified IIS-responsive transcripts that were either gonad-specific or non-gonadal. Expression datasets were further analyzed using self-organizing maps (SOMs) with hierarchical clustering. Gene network construction, functional enrichment, transcription factor enrichment, and conservation analyses were performed, and differential expression profiles were integrated with publicly available germline-, gamete-, and somatic tissue-enriched datasets.</p><p><strong>Results: </strong>Temporal transcriptomic profiling revealed distinct IIS-dependent expression trajectories in gonadal versus non-gonadal datasets. SOM-based clustering resolved temporally regulated expression modules, while network and enrichment analyses uncovered a multilayered regulatory architecture within the gonad. Gonadal expression was enriched for structural, extracellular matrix, and signaling pathway genes, whereas non-gonadal data showed enrichment for stress response and longevity-associated pathways. Integration with germline-, gamete-, and somatic tissue-enriched datasets distinguished tissue-specific regulatory signatures. Importantly, IIS-regulated gonadal components included genes highly conserved with human orthologs.</p><p><strong>Conclusion: </strong>This study provides a high-resolution temporal map of the gonadal transcriptome under reduced IIS and highlights gene modules potentially critical for reproductive maintenance. These findings offer a resource for dissecting tissue-specific aging programs and insulin-dependent regulation of reproductive health.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"23 1","pages":"529"},"PeriodicalIF":8.2,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12706931/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145764353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tetrabenazine-induced miR-34a-5p suppresses the tumorigenicity of radioresistant colorectal cancer by inhibiting M2 macrophage polarization. 四苯那嗪诱导的miR-34a-5p通过抑制M2巨噬细胞极化抑制放射耐药结直肠癌的致瘤性。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-15 DOI: 10.1186/s12964-025-02595-0
Dong Hyeon Lee, Hyun Jeong Seok, Jae Yeon Choi, Junhye Kwon, Ui Sup Shin, In Hwa Bae

Background: Radiotherapy is a very common treatment method for various cancers; however, it is not effective for patients with radioresistance. Accordingly, the discovery of drugs for patients with radioresistance cancer is critical. This study used a Food and Drug Administration (FDA)-approved drug library to identify candidate drugs for the treatment of radioresistant colorectal cancer (CRC). This approach to drug development benefits from its low cost and time requirements and can lead to rapid clinical translation.

Methods: Drugs that suppress radioresistance in CRC cells were screened. Effects of candidate drugs on cell viability and the expression of epithelial-mesenchymal transition (EMT)-related factors were evaluated through using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and quantitative real-time PCR (qRT-PCR). The effects of TBZ, identified as a candidate, on radioresistant CRC cells and organoids were evaluated using MTT, qRT-PCR, western blotting, migration, and invasion assays. Factors mediating the suppressive effects of TBZ on tumorigenicity, including the roles of Snail and the p53-miR-34a-5p axis, were evaluated using chromatin immunoprecipitation (ChIP), promoter luciferase, western blotting, qRT-PCR, and dual luciferase assays. Expression of M2 markers in THP-1-derived macrophages was confirmed by qRT-PCR.

Results: TBZ, an anti-hyperkinesia drug, was identified as a candidate agent able to reduce tumorigenicity. In radioresistant CRC cells, treatment with TBZ downregulated EMT-related factors and decreased cell migratory ability and invasiveness via reductions in Snail expression through p53-induced miR-34a-5p. Cell migration and invasion assays confirmed that TBZ had greater inhibitory effects on the migration and invasiveness of radioresistant CRC cells than those of 5-FU. Furthermore, TBZ-induced miR-34a-5p reduced M2 macrophage polarization and IL-10 secretion, thereby reducing the tumorigenicity of radioresistant CRC cells. These findings were verified using samples from patients with CRC.

Conclusions: TBZ reduces tumorigenicity via the regulation of the p53-miR-34a-5p/Snail axis in radioresistant CRC cells and suppression of M2 macrophage polarization, decreasing IL-10 secretion. Our study provides insight into drug repurposing by revealing the mechanism by which TBZ, an FDA-approved drug, reduces tumorigenicity through communication between radioresistant CRC cells and macrophages.

背景:放射治疗是各种癌症的一种非常常见的治疗方法;然而,它对有放射耐药的患者无效。因此,发现治疗放射耐药癌症的药物至关重要。本研究使用美国食品和药物管理局(FDA)批准的药物库来确定治疗放射耐药结直肠癌(CRC)的候选药物。这种方法的药物开发受益于其低成本和时间要求,并可以导致快速临床转化。方法:筛选抑制结直肠癌细胞放射耐药的药物。采用3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四唑(MTT)检测和实时荧光定量PCR (qRT-PCR)技术,评价候选药物对细胞活力和上皮间质转化(EMT)相关因子表达的影响。通过MTT、qRT-PCR、western blotting、迁移和侵袭试验评估TBZ对CRC放射耐药细胞和类器官的影响。通过染色质免疫沉淀(ChIP)、启动子荧光素酶、western blotting、qRT-PCR和双荧光素酶测定来评估介导TBZ对致瘤性抑制作用的因素,包括Snail和p53-miR-34a-5p轴的作用。qRT-PCR证实thp -1源性巨噬细胞中M2标志物的表达。结果:抗运动亢进药物TBZ是一种降低肿瘤致瘤性的候选药物。在放射耐药的CRC细胞中,TBZ治疗通过p53诱导的miR-34a-5p降低Snail表达,从而下调emt相关因子,降低细胞迁移能力和侵袭性。细胞迁移和侵袭实验证实TBZ比5-FU对CRC放射耐药细胞的迁移和侵袭有更大的抑制作用。此外,tbz诱导的miR-34a-5p降低了M2巨噬细胞极化和IL-10分泌,从而降低了放射耐药CRC细胞的致瘤性。使用结直肠癌患者的样本验证了这些发现。结论:TBZ通过调节放射耐药CRC细胞中的p53-miR-34a-5p/Snail轴,抑制M2巨噬细胞极化,降低IL-10分泌,从而降低致瘤性。我们的研究通过揭示TBZ(一种fda批准的药物)通过放射耐药CRC细胞和巨噬细胞之间的交流降低致瘤性的机制,为药物重新利用提供了见解。
{"title":"Tetrabenazine-induced miR-34a-5p suppresses the tumorigenicity of radioresistant colorectal cancer by inhibiting M2 macrophage polarization.","authors":"Dong Hyeon Lee, Hyun Jeong Seok, Jae Yeon Choi, Junhye Kwon, Ui Sup Shin, In Hwa Bae","doi":"10.1186/s12964-025-02595-0","DOIUrl":"https://doi.org/10.1186/s12964-025-02595-0","url":null,"abstract":"<p><strong>Background: </strong>Radiotherapy is a very common treatment method for various cancers; however, it is not effective for patients with radioresistance. Accordingly, the discovery of drugs for patients with radioresistance cancer is critical. This study used a Food and Drug Administration (FDA)-approved drug library to identify candidate drugs for the treatment of radioresistant colorectal cancer (CRC). This approach to drug development benefits from its low cost and time requirements and can lead to rapid clinical translation.</p><p><strong>Methods: </strong>Drugs that suppress radioresistance in CRC cells were screened. Effects of candidate drugs on cell viability and the expression of epithelial-mesenchymal transition (EMT)-related factors were evaluated through using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and quantitative real-time PCR (qRT-PCR). The effects of TBZ, identified as a candidate, on radioresistant CRC cells and organoids were evaluated using MTT, qRT-PCR, western blotting, migration, and invasion assays. Factors mediating the suppressive effects of TBZ on tumorigenicity, including the roles of Snail and the p53-miR-34a-5p axis, were evaluated using chromatin immunoprecipitation (ChIP), promoter luciferase, western blotting, qRT-PCR, and dual luciferase assays. Expression of M2 markers in THP-1-derived macrophages was confirmed by qRT-PCR.</p><p><strong>Results: </strong>TBZ, an anti-hyperkinesia drug, was identified as a candidate agent able to reduce tumorigenicity. In radioresistant CRC cells, treatment with TBZ downregulated EMT-related factors and decreased cell migratory ability and invasiveness via reductions in Snail expression through p53-induced miR-34a-5p. Cell migration and invasion assays confirmed that TBZ had greater inhibitory effects on the migration and invasiveness of radioresistant CRC cells than those of 5-FU. Furthermore, TBZ-induced miR-34a-5p reduced M2 macrophage polarization and IL-10 secretion, thereby reducing the tumorigenicity of radioresistant CRC cells. These findings were verified using samples from patients with CRC.</p><p><strong>Conclusions: </strong>TBZ reduces tumorigenicity via the regulation of the p53-miR-34a-5p/Snail axis in radioresistant CRC cells and suppression of M2 macrophage polarization, decreasing IL-10 secretion. Our study provides insight into drug repurposing by revealing the mechanism by which TBZ, an FDA-approved drug, reduces tumorigenicity through communication between radioresistant CRC cells and macrophages.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145764467","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Notch signaling pathway in regulating bone and cartilage homeostasis: novel insights into the pathogenesis and therapeutics of osteoarthritis. Notch信号通路调节骨和软骨稳态:对骨关节炎发病机制和治疗的新见解。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-15 DOI: 10.1186/s12964-025-02584-3
Haixia Wang, Ming Li, Xiaodong Wang, Juanjuan Han, Xin-An Zhang

Osteoarthritis (OA) is a chronic degenerative joint disorder characterized by cartilage degradation, bone hyperplasia, and synovitis. It is a primary cause of joint pain and dysfunction globally. The increasing incidence of OA, driven by an aging population, profoundly impairs patients' quality of life while imposing a growing economic burden on societies and families. Thus, in-depth investigations into the pathogenesis of OA, along with the exploration of novel therapeutic targets and strategies, hold significant clinical implications and social value. The Notch signaling pathway, a highly conserved intercellular communication pathway, plays a pivotal regulatory role in cell proliferation, differentiation, apoptosis, and organogenesis. In recent years, a growing body of research has revealed that the Notch signaling pathway is crucial for maintaining bone and cartilage homeostasis, with its aberrant activation or inhibition being closely linked to the initiation and progression of OA. Therefore, this narrative review performed an extensive PubMed database search using keywords like "Notch", "osteoarthritis", "bone", "cartilage", "synovitis", "osteoblasts", "osteoclasts", and "chondrocytes", and reviewed all pertinent literature. It specifically focuses on the role of Notch signaling in the differentiation and function of osteoblasts, osteoclasts, and chondrocytes, shedding light on its mechanism in cartilage damage, subchondral bone dysfunction, and synovitis. It also explores evidence for targeted Notch pathway therapies in OA, aiming to illuminate the molecular mechanisms underlying OA pathogenesis and offer new theoretical insights and therapeutic targets for OA prevention and treatment. Additionally, this narrative review seeks to decipher the mechanisms underlying the context-dependent duality of Notch signaling in bone and cartilage, and provides a critical appraisal of the challenges confronting current targeted therapies.

骨关节炎(OA)是一种慢性退行性关节疾病,以软骨退化、骨增生和滑膜炎为特征。它是全球关节疼痛和功能障碍的主要原因。在人口老龄化的推动下,OA发病率不断上升,严重影响了患者的生活质量,同时给社会和家庭带来了越来越大的经济负担。因此,深入研究OA发病机制,探索新的治疗靶点和策略,具有重要的临床意义和社会价值。Notch信号通路是一个高度保守的细胞间通讯通路,在细胞增殖、分化、凋亡和器官发生等过程中起着关键的调控作用。近年来,越来越多的研究表明Notch信号通路对维持骨和软骨稳态至关重要,其异常激活或抑制与OA的发生和发展密切相关。因此,本文使用关键词“Notch”、“骨关节炎”、“骨”、“软骨”、“滑膜炎”、“成骨细胞”、“破骨细胞”和“软骨细胞”进行了广泛的PubMed数据库搜索,并查阅了所有相关文献。重点关注Notch信号在成骨细胞、破骨细胞和软骨细胞的分化和功能中的作用,揭示其在软骨损伤、软骨下骨功能障碍和滑膜炎中的机制。探索Notch通路在OA中的靶向治疗证据,旨在阐明OA发病机制的分子机制,为OA的防治提供新的理论见解和治疗靶点。此外,这篇叙述性综述试图解读骨和软骨中Notch信号的上下文依赖性二元性的机制,并对当前靶向治疗面临的挑战进行批判性评估。
{"title":"The Notch signaling pathway in regulating bone and cartilage homeostasis: novel insights into the pathogenesis and therapeutics of osteoarthritis.","authors":"Haixia Wang, Ming Li, Xiaodong Wang, Juanjuan Han, Xin-An Zhang","doi":"10.1186/s12964-025-02584-3","DOIUrl":"https://doi.org/10.1186/s12964-025-02584-3","url":null,"abstract":"<p><p>Osteoarthritis (OA) is a chronic degenerative joint disorder characterized by cartilage degradation, bone hyperplasia, and synovitis. It is a primary cause of joint pain and dysfunction globally. The increasing incidence of OA, driven by an aging population, profoundly impairs patients' quality of life while imposing a growing economic burden on societies and families. Thus, in-depth investigations into the pathogenesis of OA, along with the exploration of novel therapeutic targets and strategies, hold significant clinical implications and social value. The Notch signaling pathway, a highly conserved intercellular communication pathway, plays a pivotal regulatory role in cell proliferation, differentiation, apoptosis, and organogenesis. In recent years, a growing body of research has revealed that the Notch signaling pathway is crucial for maintaining bone and cartilage homeostasis, with its aberrant activation or inhibition being closely linked to the initiation and progression of OA. Therefore, this narrative review performed an extensive PubMed database search using keywords like \"Notch\", \"osteoarthritis\", \"bone\", \"cartilage\", \"synovitis\", \"osteoblasts\", \"osteoclasts\", and \"chondrocytes\", and reviewed all pertinent literature. It specifically focuses on the role of Notch signaling in the differentiation and function of osteoblasts, osteoclasts, and chondrocytes, shedding light on its mechanism in cartilage damage, subchondral bone dysfunction, and synovitis. It also explores evidence for targeted Notch pathway therapies in OA, aiming to illuminate the molecular mechanisms underlying OA pathogenesis and offer new theoretical insights and therapeutic targets for OA prevention and treatment. Additionally, this narrative review seeks to decipher the mechanisms underlying the context-dependent duality of Notch signaling in bone and cartilage, and provides a critical appraisal of the challenges confronting current targeted therapies.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145764447","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cell Communication and Signaling
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
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