首页 > 最新文献

Cellular Oncology最新文献

英文 中文
Enhanced ZBTB10 expression induced by betulinic acid inhibits gastric cancer progression by inactivating the ARRDC3/ITGB4/PI3K/AKT pathway.
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2025-01-28 DOI: 10.1007/s13402-025-01039-8
Zhixin Huang, Ying Li, Zeyu Zhao, Linying Ye, Tianhao Zhang, Zihan Yu, Ertao Zhai, Yan Qian, Xiang Xu, Risheng Zhao, Shirong Cai, Jianhui Chen

Background: Gastric cancer (GC) ranks as the fourth leading cause of cancer-related deaths worldwide, with most patients diagnosed at advanced stages due to the absence of reliable early detection biomarkers.

Methods: RNA-sequencing was conducted to identify the differentially expressed genes between GC tissues and adjacent normal tissues. CCK8, EdU, colony formation, transwell, flow cytometry and xenograft assays were adopted to explore the biological function of ZBTB10 and betulinic acid (BA) in GC progression. RNA-sequencing and phospho-proteomic profiling were performed to analyze the signaling pathways associated with ZBTB10-inhibiting GC progression. Chromatin immunoprecipitation, Co-immunoprecipitation and luciferase reporter assay were employed to elucidate the potential molecular regulatory mechanisms of ZBTB10 in GC.

Results: ZBTB10 was one of the most significantly downregulated genes in GC tissues, and higher expression levels of ZBTB10 was correlated with better prognosis in patients with GC. Functional studies revealed that ZBTB10 overexpression and BA inhibited GC progression both in vitro and in vivo. Mechanistically, ZBTB10 enhanced ARRDC3 expression by binding to a specific response element in the ARRDC3 promoter region. Elevated ARRDC3 then directly interacted with β-4 integrin (ITGB4), leading to its ubiquitination and degradation. This cascade ultimately resulted in the downregulation of PI3K and AKT phosphorylation level. Moreover, ZBTB10 was a key target for BA in GC and BA inhibited GC progression through regulating the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis.

Conclusions: Our findings reveal that BA holds promise as an effective therapeutic strategy for GC, and the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis may serve as a novel diagnostic and therapeutic target.

{"title":"Enhanced ZBTB10 expression induced by betulinic acid inhibits gastric cancer progression by inactivating the ARRDC3/ITGB4/PI3K/AKT pathway.","authors":"Zhixin Huang, Ying Li, Zeyu Zhao, Linying Ye, Tianhao Zhang, Zihan Yu, Ertao Zhai, Yan Qian, Xiang Xu, Risheng Zhao, Shirong Cai, Jianhui Chen","doi":"10.1007/s13402-025-01039-8","DOIUrl":"https://doi.org/10.1007/s13402-025-01039-8","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) ranks as the fourth leading cause of cancer-related deaths worldwide, with most patients diagnosed at advanced stages due to the absence of reliable early detection biomarkers.</p><p><strong>Methods: </strong>RNA-sequencing was conducted to identify the differentially expressed genes between GC tissues and adjacent normal tissues. CCK8, EdU, colony formation, transwell, flow cytometry and xenograft assays were adopted to explore the biological function of ZBTB10 and betulinic acid (BA) in GC progression. RNA-sequencing and phospho-proteomic profiling were performed to analyze the signaling pathways associated with ZBTB10-inhibiting GC progression. Chromatin immunoprecipitation, Co-immunoprecipitation and luciferase reporter assay were employed to elucidate the potential molecular regulatory mechanisms of ZBTB10 in GC.</p><p><strong>Results: </strong>ZBTB10 was one of the most significantly downregulated genes in GC tissues, and higher expression levels of ZBTB10 was correlated with better prognosis in patients with GC. Functional studies revealed that ZBTB10 overexpression and BA inhibited GC progression both in vitro and in vivo. Mechanistically, ZBTB10 enhanced ARRDC3 expression by binding to a specific response element in the ARRDC3 promoter region. Elevated ARRDC3 then directly interacted with β-4 integrin (ITGB4), leading to its ubiquitination and degradation. This cascade ultimately resulted in the downregulation of PI3K and AKT phosphorylation level. Moreover, ZBTB10 was a key target for BA in GC and BA inhibited GC progression through regulating the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis.</p><p><strong>Conclusions: </strong>Our findings reveal that BA holds promise as an effective therapeutic strategy for GC, and the ZBTB10/ARRDC3/ITGB4/PI3K/AKT axis may serve as a novel diagnostic and therapeutic target.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143053947","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Regulatory factor X1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by transcriptional regulation of BECN1. 调节因子X1通过BECN1的转录调控促进索拉非尼诱导的肝癌铁下垂。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-09 DOI: 10.1007/s13402-024-01017-6
Zhiwen Yang, Yichuan Yuan, Yi Niu, Dinglan Zuo, Wenwu Liu, Kai Li, Yunxing Shi, Zhiyu Qiu, Keren Li, Zhu Lin, Chengrui Zhong, Zhenkun Huang, Wei He, Xinyuan Guan, Yunfei Yuan, Weian Zeng, Jiliang Qiu, Binkui Li

Background: Sorafenib is a commonly used first-line kinase-targeted drug for advanced hepatocellular carcinoma (HCC) patients suffering from limited efficacy. Emerging evidence indicates that sorafenib exerts anti-cancer activity through the induction of ferroptosis in HCC cells, but the underlying mechanism is still unclear.

Methods: The whole transcriptome sequencing and bioinformatics analysis were used to screen for target genes. The expression and subcellular localization of regulatory factor X1 (RFX1) were determined through immunohistochemistry, immunofluorescence, PCR and western blot analyses. The impact of RFX1 on HCC cell growth was assessed using CCK8, colony formation assays, cell death assays, and animal experiments. Glutathione measurement, iron assay and lipid peroxidation detection assays were performed to investigate ferroptosis of HCC cells. The regulatory mechanism of RFX1 in HCC was investigated by sgRFX1, co-IP, ChIP and luciferase experiments. Immunohistochemical and survival analyses were performed to examine the prognostic significance of RFX1 in HCC.

Results: In this study, we found that RFX1 promote ferroptosis in HCC cells. Further, we showed that sorafenib induces cell death through RFX1-mediated ferroptosis in HCC cells. The enhancing effect of RFX1 on HCC cell ferroptosis is largely dependent on inhibition of cystine/glutamate antiporter (system Xc-) activity through the BECN-SLC7A11 axis, where RFX1 directly binds to the promoter region of BECN1 and upregulates BECN1 expression. In addition, a STAT3-RFX1-BECN1 signalling loop was found to promote RFX1 expression in HCC cells.

Conclusions: Our study reveals a novel mechanism underlying sorafenib-induced HCC cell death.

背景:索拉非尼是晚期肝细胞癌(HCC)患者常用的一线激酶靶向药物,但疗效有限。新出现的证据表明索拉非尼通过诱导肝癌细胞铁下垂发挥抗癌活性,但其潜在机制尚不清楚。方法:采用全转录组测序和生物信息学分析筛选靶基因。通过免疫组织化学、免疫荧光、PCR和western blot检测调节因子X1 (RFX1)的表达和亚细胞定位。通过CCK8、菌落形成试验、细胞死亡试验和动物实验评估RFX1对HCC细胞生长的影响。采用谷胱甘肽测定、铁含量测定和脂质过氧化检测检测肝癌细胞铁下垂。通过sgRFX1、co-IP、ChIP和荧光素酶实验研究RFX1在HCC中的调控机制。通过免疫组织化学和生存分析来检验RFX1在HCC中的预后意义。结果:本研究发现RFX1促进肝癌细胞铁下垂。此外,我们发现索拉非尼通过rfx1介导的肝癌细胞铁凋亡诱导细胞死亡。RFX1对HCC细胞铁凋亡的增强作用主要依赖于通过BECN-SLC7A11轴抑制胱氨酸/谷氨酸反转运蛋白(系统Xc-)活性,其中RFX1直接结合BECN1的启动子区域并上调BECN1的表达。此外,STAT3-RFX1-BECN1信号通路在HCC细胞中促进RFX1的表达。结论:我们的研究揭示了索拉非尼诱导HCC细胞死亡的新机制。
{"title":"Regulatory factor X1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by transcriptional regulation of BECN1.","authors":"Zhiwen Yang, Yichuan Yuan, Yi Niu, Dinglan Zuo, Wenwu Liu, Kai Li, Yunxing Shi, Zhiyu Qiu, Keren Li, Zhu Lin, Chengrui Zhong, Zhenkun Huang, Wei He, Xinyuan Guan, Yunfei Yuan, Weian Zeng, Jiliang Qiu, Binkui Li","doi":"10.1007/s13402-024-01017-6","DOIUrl":"https://doi.org/10.1007/s13402-024-01017-6","url":null,"abstract":"<p><strong>Background: </strong>Sorafenib is a commonly used first-line kinase-targeted drug for advanced hepatocellular carcinoma (HCC) patients suffering from limited efficacy. Emerging evidence indicates that sorafenib exerts anti-cancer activity through the induction of ferroptosis in HCC cells, but the underlying mechanism is still unclear.</p><p><strong>Methods: </strong>The whole transcriptome sequencing and bioinformatics analysis were used to screen for target genes. The expression and subcellular localization of regulatory factor X1 (RFX1) were determined through immunohistochemistry, immunofluorescence, PCR and western blot analyses. The impact of RFX1 on HCC cell growth was assessed using CCK8, colony formation assays, cell death assays, and animal experiments. Glutathione measurement, iron assay and lipid peroxidation detection assays were performed to investigate ferroptosis of HCC cells. The regulatory mechanism of RFX1 in HCC was investigated by sgRFX1, co-IP, ChIP and luciferase experiments. Immunohistochemical and survival analyses were performed to examine the prognostic significance of RFX1 in HCC.</p><p><strong>Results: </strong>In this study, we found that RFX1 promote ferroptosis in HCC cells. Further, we showed that sorafenib induces cell death through RFX1-mediated ferroptosis in HCC cells. The enhancing effect of RFX1 on HCC cell ferroptosis is largely dependent on inhibition of cystine/glutamate antiporter (system Xc-) activity through the BECN-SLC7A11 axis, where RFX1 directly binds to the promoter region of BECN1 and upregulates BECN1 expression. In addition, a STAT3-RFX1-BECN1 signalling loop was found to promote RFX1 expression in HCC cells.</p><p><strong>Conclusions: </strong>Our study reveals a novel mechanism underlying sorafenib-induced HCC cell death.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2024-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142802480","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Senolytics: charting a new course or enhancing existing anti-tumor therapies? 老年学:制定新的治疗方案还是加强现有的抗肿瘤疗法?
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-04 DOI: 10.1007/s13402-024-01018-5
Konrad Czajkowski, Mariola Herbet, Marek Murias, Iwona Piątkowska-Chmiel

Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.

细胞衰老是机体的一种自然反应。最初,它被认为是一种有效的抗肿瘤机制。然而,现在人们认为,虽然细胞衰老最初是防止肿瘤发生的强大屏障,但随后衰老细胞的积累可能矛盾地促进癌症复发并对邻近组织造成损害。细胞增殖和衰老之间的复杂平衡在维持组织稳态中起着关键作用。此外,衰老细胞分泌许多生物活性分子,统称为衰老相关分泌表型(SASP),可诱导慢性炎症,改变组织结构,并通过旁分泌信号促进肿瘤发生。在成千上万的化合物中,老年治疗药物已经成为抗癌治疗中非常有前途的候选药物。它们选择性靶向衰老细胞,同时保留健康组织的能力代表了治疗干预的范式转变,为个性化肿瘤医学提供了新的途径。抗衰老药物通过靶向清除衰老细胞,引入了新的治疗可能性。作为单独的药物,它们可以清除处于衰老状态的肿瘤细胞,当与化疗或放疗联合使用时,可以清除治疗后残留的衰老癌细胞。这种双重方法允许有意使用低剂量治疗或去除治疗后意外的衰老细胞。此外,通过靶向非癌性衰老细胞,抗衰老药可能有助于降低肿瘤形成风险,限制复发,减缓疾病进展。本文探讨了细胞衰老的机制,它在癌症治疗中的作用,衰老治疗的重要性,特别关注抗衰老药物的治疗潜力。
{"title":"Senolytics: charting a new course or enhancing existing anti-tumor therapies?","authors":"Konrad Czajkowski, Mariola Herbet, Marek Murias, Iwona Piątkowska-Chmiel","doi":"10.1007/s13402-024-01018-5","DOIUrl":"https://doi.org/10.1007/s13402-024-01018-5","url":null,"abstract":"<p><p>Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":""},"PeriodicalIF":4.9,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142781643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GALNT6 drives lenvatinib resistance in hepatocellular carcinoma through autophagy and cancer-associated fibroblast activation. GALNT6通过自噬和癌症相关成纤维细胞激活驱动肝细胞癌对lenvatinib的耐药性。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-12-24 DOI: 10.1007/s13402-024-01032-7
Peiling Zhang, Shiping Chen, Jialiang Cai, Lina Song, Bing Quan, Jinglei Wan, Guiqi Zhu, Biao Wang, Yi Yang, Zhengjun Zhou, Tao Li, Zhi Dai

Background: Hepatocellular carcinoma (HCC) remains a significant global health challenge with limited treatment options. Lenvatinib, a tyrosine kinase inhibitor, has shown promise but is often undermined by the development of drug resistance.

Methods: Utilizing high-throughput sequencing, we investigated the molecular mechanisms underlying lenvatinib resistance in HCC cells, with a focus on metabolic pathways. Key genes, including GALNT6, were validated through quantitative real-time PCR. The effects of GALNT6 knockdown on lenvatinib sensitivity were examined in vitro and in vivo. O-GalNAc glycosylation was assessed using Vicia Villosa Lectin. Immune cell infiltration and interactions were analyzed in the TCGA-LIHC cohort, with further validation by Western blotting and immunohistochemistry.

Results: Our findings indicate that lenvatinib resistance in HCC is driven by the mucin-type O-glycosylation pathway, with GALNT6 playing a critical role. Knockdown of GALNT6 led to reduced O-GalNAc glycosylation, including the modification of LAPTM5, resulting in decreased LAPTM5 activity and autophagy inhibition. Additionally, GALNT6 silencing disrupted the PDGFA-PDGFRB axis, impairing the activation of cancer-associated fibroblasts (CAFs) and reducing their secretion of SPP1, which collectively diminished lenvatinib resistance.

Conclusions: GALNT6 is integral to the resistance mechanisms against lenvatinib in HCC by modulating autophagy and CAF activation. Targeting GALNT6 offers a promising strategy to enhance lenvatinib efficacy and improve therapeutic outcomes in HCC.

背景:肝细胞癌(HCC)仍然是一个重大的全球健康挑战,治疗方案有限。Lenvatinib是一种酪氨酸激酶抑制剂,已经显示出前景,但经常因耐药性的发展而受到损害。方法:利用高通量测序技术,研究肝癌细胞lenvatinib耐药的分子机制,重点研究代谢途径。通过实时荧光定量PCR对GALNT6等关键基因进行验证。在体外和体内研究GALNT6敲低对lenvatinib敏感性的影响。O-GalNAc糖基化评价采用紫薇凝集素。在TCGA-LIHC队列中分析免疫细胞浸润和相互作用,并通过免疫印迹和免疫组织化学进一步验证。结果:我们的研究结果表明,HCC中的lenvatinib耐药是由粘蛋白型o糖基化途径驱动的,GALNT6在其中发挥了关键作用。GALNT6的敲低导致O-GalNAc糖基化减少,包括LAPTM5的修饰,导致LAPTM5活性降低和自噬抑制。此外,GALNT6沉默破坏了PDGFA-PDGFRB轴,损害了癌症相关成纤维细胞(CAFs)的激活并减少了它们的SPP1分泌,这共同降低了lenvatinib耐药性。结论:GALNT6通过调节细胞自噬和CAF激活,在HCC中对lenvatinib的耐药机制中是不可或缺的。靶向GALNT6提供了一种有希望的策略来提高lenvatinib在HCC中的疗效和改善治疗结果。
{"title":"GALNT6 drives lenvatinib resistance in hepatocellular carcinoma through autophagy and cancer-associated fibroblast activation.","authors":"Peiling Zhang, Shiping Chen, Jialiang Cai, Lina Song, Bing Quan, Jinglei Wan, Guiqi Zhu, Biao Wang, Yi Yang, Zhengjun Zhou, Tao Li, Zhi Dai","doi":"10.1007/s13402-024-01032-7","DOIUrl":"10.1007/s13402-024-01032-7","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) remains a significant global health challenge with limited treatment options. Lenvatinib, a tyrosine kinase inhibitor, has shown promise but is often undermined by the development of drug resistance.</p><p><strong>Methods: </strong>Utilizing high-throughput sequencing, we investigated the molecular mechanisms underlying lenvatinib resistance in HCC cells, with a focus on metabolic pathways. Key genes, including GALNT6, were validated through quantitative real-time PCR. The effects of GALNT6 knockdown on lenvatinib sensitivity were examined in vitro and in vivo. O-GalNAc glycosylation was assessed using Vicia Villosa Lectin. Immune cell infiltration and interactions were analyzed in the TCGA-LIHC cohort, with further validation by Western blotting and immunohistochemistry.</p><p><strong>Results: </strong>Our findings indicate that lenvatinib resistance in HCC is driven by the mucin-type O-glycosylation pathway, with GALNT6 playing a critical role. Knockdown of GALNT6 led to reduced O-GalNAc glycosylation, including the modification of LAPTM5, resulting in decreased LAPTM5 activity and autophagy inhibition. Additionally, GALNT6 silencing disrupted the PDGFA-PDGFRB axis, impairing the activation of cancer-associated fibroblasts (CAFs) and reducing their secretion of SPP1, which collectively diminished lenvatinib resistance.</p><p><strong>Conclusions: </strong>GALNT6 is integral to the resistance mechanisms against lenvatinib in HCC by modulating autophagy and CAF activation. Targeting GALNT6 offers a promising strategy to enhance lenvatinib efficacy and improve therapeutic outcomes in HCC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2439-2460"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142883509","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Regulation of AUF1 alternative splicing by hnRNPA1 and SRSF2 modulate the sensitivity of ovarian cancer cells to cisplatin. hnRNPA1和SRSF2调控AUF1选择性剪接可调节卵巢癌细胞对顺铂的敏感性。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-12-09 DOI: 10.1007/s13402-024-01023-8
Jia-Mei Wang, Ning Liu, Xue-Jing Wei, Fu-Ying Zhao, Chao Li, Hua-Qin Wang, Chuan Liu

Purpose: Clarification of cisplatin resistance may provide new targets for therapy in cisplatin resistant ovarian cancer. The current study aims to explore involvement of isoforms of AU-rich element RNA-binding protein 1 (AUF1) in cisplatin resistance in ovarian cancer.

Methods: The cancer stem cell-like features were analyzed using colony formation assay, tumor sphere formation assay and nude mouse xenograft experiments. AUF1 isoforms expression was analyzed using immunoblotting, qRT-PCR, and immunohistochemistry. RIP and Biotin pulldown was used to analyze the interaction of SRSF2 and hnRNPA1 with AUF1 transcript. Transcriptome regulated by AUF1 isoforms was analyzed by RNA-seq.

Results: The current study demonstrated differential expression of AUF1 isoforms in cisplatin sensitive and resistant ovarian cancer tissues and cells. P37 isoform promoted proliferation, while p45 isoform enhanced responsiveness of ovarian cancer cells to cisplatin. the clonal formation capacity of the cells, and the restoration of p45 expression reduced the capacity with cisplatin treatment. The competitive binding of phosphorylated hnRNPA1 and O-GlcNAc-modified SRSF2 on AUF1 exon 2 and exon 7 regulated the alternative splicing of AUF1.

Conclusion: The competitive binding of phosphorylated hnRNPA1 and O-GlcNAc modified SRSF2 on exon 2 and exon 7 regulated the alternative splicing of AUF1 and subsequent isoform expression. P37 isoform played a "cancer promoter" role, p42 and p45, especially p45 played a "cancer suppressor" role in ovarian cancer. This study provides a new target for exploring the drug resistance mechanism of ovarian cancer.

目的:明确顺铂耐药情况,为顺铂耐药卵巢癌的治疗提供新的靶点。本研究旨在探讨富au元素rna结合蛋白1 (AUF1)亚型在卵巢癌顺铂耐药中的作用。方法:采用集落形成实验、肿瘤球形成实验和裸鼠异种移植实验分析肿瘤干细胞样特征。使用免疫印迹、qRT-PCR和免疫组织化学分析AUF1亚型的表达。利用RIP和Biotin pulldown分析SRSF2和hnRNPA1与AUF1转录物的相互作用。通过RNA-seq分析AUF1亚型调控的转录组。结果:目前的研究表明,AUF1亚型在顺铂敏感和耐药的卵巢癌组织和细胞中存在差异表达。P37异构体促进卵巢癌细胞增殖,而p45异构体增强卵巢癌细胞对顺铂的反应性。顺铂治疗降低了细胞的克隆形成能力和p45表达的恢复。磷酸化hnRNPA1和o - glcnac修饰的SRSF2在AUF1外显子2和外显子7上的竞争性结合调节了AUF1的选择性剪接。结论:磷酸化hnRNPA1和O-GlcNAc修饰的SRSF2在外显子2和外显子7上的竞争性结合调节了AUF1的选择性剪接和随后的异构体表达。P37异构体在卵巢癌中起“癌启动子”作用,p42和p45,尤其是p45起“癌抑制子”作用。本研究为探索卵巢癌耐药机制提供了新的靶点。
{"title":"Regulation of AUF1 alternative splicing by hnRNPA1 and SRSF2 modulate the sensitivity of ovarian cancer cells to cisplatin.","authors":"Jia-Mei Wang, Ning Liu, Xue-Jing Wei, Fu-Ying Zhao, Chao Li, Hua-Qin Wang, Chuan Liu","doi":"10.1007/s13402-024-01023-8","DOIUrl":"10.1007/s13402-024-01023-8","url":null,"abstract":"<p><strong>Purpose: </strong>Clarification of cisplatin resistance may provide new targets for therapy in cisplatin resistant ovarian cancer. The current study aims to explore involvement of isoforms of AU-rich element RNA-binding protein 1 (AUF1) in cisplatin resistance in ovarian cancer.</p><p><strong>Methods: </strong>The cancer stem cell-like features were analyzed using colony formation assay, tumor sphere formation assay and nude mouse xenograft experiments. AUF1 isoforms expression was analyzed using immunoblotting, qRT-PCR, and immunohistochemistry. RIP and Biotin pulldown was used to analyze the interaction of SRSF2 and hnRNPA1 with AUF1 transcript. Transcriptome regulated by AUF1 isoforms was analyzed by RNA-seq.</p><p><strong>Results: </strong>The current study demonstrated differential expression of AUF1 isoforms in cisplatin sensitive and resistant ovarian cancer tissues and cells. P37 isoform promoted proliferation, while p45 isoform enhanced responsiveness of ovarian cancer cells to cisplatin. the clonal formation capacity of the cells, and the restoration of p45 expression reduced the capacity with cisplatin treatment. The competitive binding of phosphorylated hnRNPA1 and O-GlcNAc-modified SRSF2 on AUF1 exon 2 and exon 7 regulated the alternative splicing of AUF1.</p><p><strong>Conclusion: </strong>The competitive binding of phosphorylated hnRNPA1 and O-GlcNAc modified SRSF2 on exon 2 and exon 7 regulated the alternative splicing of AUF1 and subsequent isoform expression. P37 isoform played a \"cancer promoter\" role, p42 and p45, especially p45 played a \"cancer suppressor\" role in ovarian cancer. This study provides a new target for exploring the drug resistance mechanism of ovarian cancer.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2349-2366"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142802466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring tumor microenvironment interactions and apoptosis pathways in NSCLC through spatial transcriptomics and machine learning. 通过空间转录组学和机器学习探索非小细胞肺癌的肿瘤微环境相互作用和凋亡途径。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-12-19 DOI: 10.1007/s13402-024-01025-6
Huimin Li, Yuheng Jiao, Yi Zhang, Junzhi Liu, Shuixian Huang

Background: The most common type of lung cancer is non-small cell lung cancer (NSCLC), accounting for 85% of all cases. Programmed cell death (PCD), an important regulatory mechanism for cell survival and homeostasis, has become increasingly prominent in cancer research in recent years. As such, exploring the role of PCD in NSCLC may help uncover new mechanisms for therapeutic targets.

Methods: We utilized the GEO database and TCGA NSCLC gene data to screen for co-expressed genes. To delve deeper, single-cell sequencing combined with spatial transcriptomics was employed to study the intrinsic mechanisms of programmed cell death in cells and their interaction with the tumor microenvironment. Furthermore, Mendelian randomization was applied to screen for causally related genes. Prognostic models were constructed using various machine learning algorithms, and multi-cohort multi-omics analyses were conducted to screen for genes. In vitro experiments were then carried out to reveal the biological functions of the genes and their relationship with apoptosis.

Results: Cells with high programmed cell death activity primarily activate pathways related to apoptosis, cell migration, and hypoxia, while also exhibiting strong interactions with smooth muscle cells in the tumor microenvironment. Based on a set of programmed cell death genes, the prognostic model NSCLCPCD demonstrates strong predictive capabilities. Moreover, laboratory experiments confirm that SLC7A5 promotes the proliferation of NSCLC cells, and the knockout of SLC7A5 significantly increases tumor cell apoptosis.

Conclusions: Our data indicate that programmed cell death is predominantly associated with pathways related to apoptosis, tumor metastasis, and hypoxia. Additionally, it suggests that SLC7A5 is a significant risk indicator for the prognosis of non-small cell lung cancer (NSCLC) and may serve as an effective target for enhancing apoptosis in NSCLC tumor cells.

背景:最常见的肺癌类型是非小细胞肺癌(NSCLC),占所有病例的85%。细胞程序性死亡(Programmed cell death, PCD)作为细胞存活和稳态的重要调控机制,近年来在肿瘤研究中得到越来越多的关注。因此,探索PCD在非小细胞肺癌中的作用可能有助于揭示治疗靶点的新机制。方法:利用GEO数据库和TCGA NSCLC基因数据筛选共表达基因。为了深入研究,我们采用单细胞测序结合空间转录组学来研究细胞程序性死亡的内在机制及其与肿瘤微环境的相互作用。此外,孟德尔随机化应用筛选因果相关基因。使用各种机器学习算法构建预后模型,并进行多队列多组学分析以筛选基因。然后进行体外实验,揭示基因的生物学功能及其与细胞凋亡的关系。结果:具有高程序性细胞死亡活性的细胞主要激活与凋亡、细胞迁移和缺氧相关的途径,同时也与肿瘤微环境中的平滑肌细胞表现出强烈的相互作用。基于一组程序性细胞死亡基因,预后模型NSCLCPCD显示出强大的预测能力。此外,实验室实验证实SLC7A5促进了NSCLC细胞的增殖,敲除SLC7A5可显著增加肿瘤细胞的凋亡。结论:我们的数据表明程序性细胞死亡主要与凋亡、肿瘤转移和缺氧相关的途径相关。提示SLC7A5是非小细胞肺癌(non-small cell lung cancer, NSCLC)预后的重要危险指标,可能是促进NSCLC肿瘤细胞凋亡的有效靶点。
{"title":"Exploring tumor microenvironment interactions and apoptosis pathways in NSCLC through spatial transcriptomics and machine learning.","authors":"Huimin Li, Yuheng Jiao, Yi Zhang, Junzhi Liu, Shuixian Huang","doi":"10.1007/s13402-024-01025-6","DOIUrl":"10.1007/s13402-024-01025-6","url":null,"abstract":"<p><strong>Background: </strong>The most common type of lung cancer is non-small cell lung cancer (NSCLC), accounting for 85% of all cases. Programmed cell death (PCD), an important regulatory mechanism for cell survival and homeostasis, has become increasingly prominent in cancer research in recent years. As such, exploring the role of PCD in NSCLC may help uncover new mechanisms for therapeutic targets.</p><p><strong>Methods: </strong>We utilized the GEO database and TCGA NSCLC gene data to screen for co-expressed genes. To delve deeper, single-cell sequencing combined with spatial transcriptomics was employed to study the intrinsic mechanisms of programmed cell death in cells and their interaction with the tumor microenvironment. Furthermore, Mendelian randomization was applied to screen for causally related genes. Prognostic models were constructed using various machine learning algorithms, and multi-cohort multi-omics analyses were conducted to screen for genes. In vitro experiments were then carried out to reveal the biological functions of the genes and their relationship with apoptosis.</p><p><strong>Results: </strong>Cells with high programmed cell death activity primarily activate pathways related to apoptosis, cell migration, and hypoxia, while also exhibiting strong interactions with smooth muscle cells in the tumor microenvironment. Based on a set of programmed cell death genes, the prognostic model NSCLCPCD demonstrates strong predictive capabilities. Moreover, laboratory experiments confirm that SLC7A5 promotes the proliferation of NSCLC cells, and the knockout of SLC7A5 significantly increases tumor cell apoptosis.</p><p><strong>Conclusions: </strong>Our data indicate that programmed cell death is predominantly associated with pathways related to apoptosis, tumor metastasis, and hypoxia. Additionally, it suggests that SLC7A5 is a significant risk indicator for the prognosis of non-small cell lung cancer (NSCLC) and may serve as an effective target for enhancing apoptosis in NSCLC tumor cells.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2383-2405"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142856122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of BRCA1 in glioblastoma etiology. BRCA1在胶质母细胞瘤发病中的作用。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-12-10 DOI: 10.1007/s13402-024-01024-7
Emirhan Harbi, Michael Aschner

BRCA1 (Breast Cancer 1) is a tumor suppressor gene with a role in DNA repair by Homologous Recombination (HR), and maintenance of genomic stability that is frequently investigated in breast, prostate, and ovarian cancers. BRCA1 mutations or dysregulation in glioblastoma can lead to impaired DNA repair mechanisms, resulting in tumor progression and resistance to standard therapies. Several studies have shown that BRCA1 expression is altered, albeit rarely, in glioblastoma, leading to poor prognosis and increased tumor aggressiveness. In addition, the communication of BRCA1 with other molecular pathways such as p53 and PTEN further complicates its role in glioblastoma pathogenesis. Targeting BRCA1-related pathways in these cases has shown the potential to improve the efficacy of standard treatments, including radiotherapy and chemotherapy. The development of (Poly (ADP-ribose) Polymerase) PARP inhibitors that exploit the lack of HR also offers a therapeutic approach to glioblastoma patients with BRCA1 mutations. Despite these advances, the heterogeneity of glioblastoma and its complex tumor microenvironment make the translation of such approaches into clinical practice still challenging, and there is an "unmet need". This review discusses the current mechanisms of etiology and potential treatment of BRCA1-related glioblastoma.

BRCA1 (Breast Cancer 1)是一种肿瘤抑制基因,在通过同源重组(Homologous Recombination, HR)修复DNA和维持基因组稳定性中发挥作用,在乳腺癌、前列腺癌和卵巢癌中经常被研究。胶质母细胞瘤中的BRCA1突变或失调可导致DNA修复机制受损,导致肿瘤进展和对标准治疗的耐药性。几项研究表明,BRCA1表达在胶质母细胞瘤中发生改变(尽管很少),导致预后不良和肿瘤侵袭性增加。此外,BRCA1与p53、PTEN等其他分子通路的通讯使其在胶质母细胞瘤发病中的作用进一步复杂化。在这些病例中,靶向brca1相关通路已显示出提高标准治疗(包括放疗和化疗)疗效的潜力。利用HR缺乏的聚(adp核糖)聚合酶PARP抑制剂的开发也为BRCA1突变的胶质母细胞瘤患者提供了一种治疗方法。尽管取得了这些进展,但胶质母细胞瘤的异质性及其复杂的肿瘤微环境使得这些方法转化为临床实践仍然具有挑战性,并且存在“未满足的需求”。本文综述了brca1相关胶质母细胞瘤的病因机制和潜在治疗方法。
{"title":"Role of BRCA1 in glioblastoma etiology.","authors":"Emirhan Harbi, Michael Aschner","doi":"10.1007/s13402-024-01024-7","DOIUrl":"10.1007/s13402-024-01024-7","url":null,"abstract":"<p><p>BRCA1 (Breast Cancer 1) is a tumor suppressor gene with a role in DNA repair by Homologous Recombination (HR), and maintenance of genomic stability that is frequently investigated in breast, prostate, and ovarian cancers. BRCA1 mutations or dysregulation in glioblastoma can lead to impaired DNA repair mechanisms, resulting in tumor progression and resistance to standard therapies. Several studies have shown that BRCA1 expression is altered, albeit rarely, in glioblastoma, leading to poor prognosis and increased tumor aggressiveness. In addition, the communication of BRCA1 with other molecular pathways such as p53 and PTEN further complicates its role in glioblastoma pathogenesis. Targeting BRCA1-related pathways in these cases has shown the potential to improve the efficacy of standard treatments, including radiotherapy and chemotherapy. The development of (Poly (ADP-ribose) Polymerase) PARP inhibitors that exploit the lack of HR also offers a therapeutic approach to glioblastoma patients with BRCA1 mutations. Despite these advances, the heterogeneity of glioblastoma and its complex tumor microenvironment make the translation of such approaches into clinical practice still challenging, and there is an \"unmet need\". This review discusses the current mechanisms of etiology and potential treatment of BRCA1-related glioblastoma.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2091-2098"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142802777","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
RAB37 suppresses the EMT, migration and invasion of gastric cancer cells by mediating autophagic degradation of β-catenin. RAB37通过介导β-catenin的自噬降解,抑制胃癌细胞的EMT、迁移和侵袭。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-12-19 DOI: 10.1007/s13402-024-01028-3
Jiangling Duan, Xiuyin Guan, Jiaxin Xue, Jiayu Wang, Zhiwei Wang, Xuan Chen, Wen Jiang, Wannian Sui, Yongfang Song, Tianshu Li, Dewang Rao, Xueyan Wu, Ming Lu

Background: Gastric cancer, characterized by its high morbidity and mortality rates, exhibits low levels of RAB37. The role and molecular mechanisms of RAB37, a small GTPase, in the pathogenesis of gastric cancer are still unclear.

Methods: We assessed RAB37 expression in gastric cancer cells using quantitative Polymerase Chain Reaction (qPCR), Western blot, and immunohistochemical staining (IHC), and analyzed EMT marker proteins and autophagy changes via Western blot, immunofluorescence (IF), and transmission electron microscopy (TEM). Co-immunoprecipitation (co-IP) was used to identify protein-protein interactions. We studied the migration and invasion of gastric cancer cells using wound healing and transwell assays in vitro and a mouse pulmonary metastasis model in vivo.

Results: Overexpression of RAB37 suppressed EMT, invasion, and migration while enhancing autophagy in gastric cancer cells, which was dependent on its GTPase activity. However, all these effects could be reversed by the autophagy inhibitor chloroquine. Regarding the molecular mechanism, RAB37 strengthened the interaction between p62 and β-catenin, which consequently enhanced the p62-mediated autophagic degradation of β-catenin. Furthermore, RAB37 curbed the pulmonary metastasis of both general and cisplatin-resistant gastric cancer cells.

Conclusion: The low level of RAB37 reduces interaction between p62 and β-catenin and then the autophagic degradation of β-catenin, thereby promoting the EMT, invasion, and migration in gastric cancer cells. The low expression of RAB37 in gastric cancer suggests a potential therapeutic target, especially for cisplatin-resistant gastric cancer.

背景:胃癌具有高发病率和高死亡率的特点,RAB37水平较低。RAB37是一种小的GTPase,其在胃癌发病中的作用和分子机制尚不清楚。方法:采用定量聚合酶链式反应(qPCR)、免疫印迹(Western blot)和免疫组化染色(IHC)检测RAB37在胃癌细胞中的表达,并通过免疫印迹(Western blot)、免疫荧光(IF)和透射电镜(TEM)分析EMT标记蛋白和自噬的变化。共免疫沉淀(co-IP)用于鉴定蛋白质之间的相互作用。我们在体外通过伤口愈合和transwell实验以及小鼠肺转移模型研究了胃癌细胞的迁移和侵袭。结果:RAB37过表达抑制胃癌细胞的EMT、侵袭和迁移,同时增强胃癌细胞的自噬,其作用机制依赖于RAB37的GTPase活性。然而,所有这些作用都可以被自噬抑制剂氯喹逆转。在分子机制上,RAB37增强了p62与β-catenin的相互作用,从而增强了p62介导的β-catenin的自噬降解。此外,RAB37抑制了一般和顺铂耐药胃癌细胞的肺转移。结论:低水平的RAB37降低了p62与β-catenin的相互作用,进而降低了β-catenin的自噬降解,从而促进了胃癌细胞的EMT、侵袭和迁移。RAB37在胃癌中的低表达提示了一个潜在的治疗靶点,特别是对顺铂耐药的胃癌。
{"title":"RAB37 suppresses the EMT, migration and invasion of gastric cancer cells by mediating autophagic degradation of β-catenin.","authors":"Jiangling Duan, Xiuyin Guan, Jiaxin Xue, Jiayu Wang, Zhiwei Wang, Xuan Chen, Wen Jiang, Wannian Sui, Yongfang Song, Tianshu Li, Dewang Rao, Xueyan Wu, Ming Lu","doi":"10.1007/s13402-024-01028-3","DOIUrl":"10.1007/s13402-024-01028-3","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer, characterized by its high morbidity and mortality rates, exhibits low levels of RAB37. The role and molecular mechanisms of RAB37, a small GTPase, in the pathogenesis of gastric cancer are still unclear.</p><p><strong>Methods: </strong>We assessed RAB37 expression in gastric cancer cells using quantitative Polymerase Chain Reaction (qPCR), Western blot, and immunohistochemical staining (IHC), and analyzed EMT marker proteins and autophagy changes via Western blot, immunofluorescence (IF), and transmission electron microscopy (TEM). Co-immunoprecipitation (co-IP) was used to identify protein-protein interactions. We studied the migration and invasion of gastric cancer cells using wound healing and transwell assays in vitro and a mouse pulmonary metastasis model in vivo.</p><p><strong>Results: </strong>Overexpression of RAB37 suppressed EMT, invasion, and migration while enhancing autophagy in gastric cancer cells, which was dependent on its GTPase activity. However, all these effects could be reversed by the autophagy inhibitor chloroquine. Regarding the molecular mechanism, RAB37 strengthened the interaction between p62 and β-catenin, which consequently enhanced the p62-mediated autophagic degradation of β-catenin. Furthermore, RAB37 curbed the pulmonary metastasis of both general and cisplatin-resistant gastric cancer cells.</p><p><strong>Conclusion: </strong>The low level of RAB37 reduces interaction between p62 and β-catenin and then the autophagic degradation of β-catenin, thereby promoting the EMT, invasion, and migration in gastric cancer cells. The low expression of RAB37 in gastric cancer suggests a potential therapeutic target, especially for cisplatin-resistant gastric cancer.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2407-2421"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142856227","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Non-canonical function of PHGDH promotes HCC metastasis by interacting with METTL3. 非典型功能的PHGDH通过与METTL3相互作用促进HCC转移。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-12-18 DOI: 10.1007/s13402-024-01029-2
Bin Cheng, Jing Ma, Ni Tang, Rui Liu, Pai Peng, Kai Wang

Purpose: Phosphoglycerate dehydrogenase (PHGDH), a pivotal enzyme in serine synthesis, plays a key role in the malignant progression of tumors through both its metabolic activity and moonlight functions. This study aims to elucidate the non-canonical function of PHGDH in promoting hepatocellular carcinoma (HCC) metastasis through its interaction with methyltransferase-like 3 (METTL3), potentially uncovering a novel therapeutic target.

Methods: Western blot was used to study PHGDH expression changes under anoikis and cellular functional assays were employed to assess its role in HCC metastasis. PHGDH-METTL3 interactions were explored using GST pull-down, Co-immunoprecipitation and immunofluorescence assays. Protein stability and ubiquitination assays were performed to understand PHGDH's impact on METTL3. Flow cytometry, cellular assays and nude mice model were used to confirm PHGDH's effects on anoikis resistance and HCC metastasis in vitro and in vivo.

Results: PHGDH is upregulated under anoikis conditions, thereby enhancing the metastatic potential of HCC cells. By interacting with METTL3, PHGDH prevents its ubiquitin-dependent degradation, resulting in higher METTL3 protein levels. This interaction upregulates epithelial-mesenchymal transition related genes, contributing to anoikis resistance and HCC metastasis. Nude mice model confirms that PHGDH's interaction with METTL3 is crucial for driving HCC metastasis.

Conclusion: Our research presents the first evidence that PHGDH promotes HCC metastasis by interacting with METTL3. The PHGDH-METTL3 axis may serve as a potential clinical therapeutic target, offering new insights into the multifaceted roles of PHGDH in HCC metastasis.

目的:磷酸甘油酸脱氢酶(Phosphoglycerate dehydrogenase, PHGDH)是丝氨酸合成的关键酶,其代谢活性和月相功能在肿瘤的恶性发展中起着关键作用。本研究旨在阐明PHGDH通过与甲基转移酶样3 (METTL3)的相互作用促进肝细胞癌(HCC)转移的非典型功能,潜在地发现一个新的治疗靶点。方法:采用Western blot法检测肝癌细胞中PHGDH的表达变化,采用细胞功能测定法评价其在肝癌转移中的作用。利用GST下拉法、共免疫沉淀法和免疫荧光法研究PHGDH-METTL3相互作用。通过蛋白稳定性和泛素化分析来了解PHGDH对METTL3的影响。通过流式细胞术、细胞实验和裸鼠模型验证了PHGDH在体外和体内对肝癌耐药和肝癌转移的影响。结果:PHGDH在anoikis条件下上调,从而增强HCC细胞的转移潜能。通过与METTL3相互作用,PHGDH阻止其泛素依赖性降解,导致更高的METTL3蛋白水平。这种相互作用上调了上皮-间质转化相关基因,促进了anoikis耐药性和HCC转移。裸鼠模型证实了PHGDH与METTL3的相互作用对HCC转移至关重要。结论:我们的研究首次提供了PHGDH通过与METTL3相互作用促进HCC转移的证据。PHGDH- mettl3轴可能作为一个潜在的临床治疗靶点,为PHGDH在HCC转移中的多方面作用提供了新的见解。
{"title":"Non-canonical function of PHGDH promotes HCC metastasis by interacting with METTL3.","authors":"Bin Cheng, Jing Ma, Ni Tang, Rui Liu, Pai Peng, Kai Wang","doi":"10.1007/s13402-024-01029-2","DOIUrl":"10.1007/s13402-024-01029-2","url":null,"abstract":"<p><strong>Purpose: </strong>Phosphoglycerate dehydrogenase (PHGDH), a pivotal enzyme in serine synthesis, plays a key role in the malignant progression of tumors through both its metabolic activity and moonlight functions. This study aims to elucidate the non-canonical function of PHGDH in promoting hepatocellular carcinoma (HCC) metastasis through its interaction with methyltransferase-like 3 (METTL3), potentially uncovering a novel therapeutic target.</p><p><strong>Methods: </strong>Western blot was used to study PHGDH expression changes under anoikis and cellular functional assays were employed to assess its role in HCC metastasis. PHGDH-METTL3 interactions were explored using GST pull-down, Co-immunoprecipitation and immunofluorescence assays. Protein stability and ubiquitination assays were performed to understand PHGDH's impact on METTL3. Flow cytometry, cellular assays and nude mice model were used to confirm PHGDH's effects on anoikis resistance and HCC metastasis in vitro and in vivo.</p><p><strong>Results: </strong>PHGDH is upregulated under anoikis conditions, thereby enhancing the metastatic potential of HCC cells. By interacting with METTL3, PHGDH prevents its ubiquitin-dependent degradation, resulting in higher METTL3 protein levels. This interaction upregulates epithelial-mesenchymal transition related genes, contributing to anoikis resistance and HCC metastasis. Nude mice model confirms that PHGDH's interaction with METTL3 is crucial for driving HCC metastasis.</p><p><strong>Conclusion: </strong>Our research presents the first evidence that PHGDH promotes HCC metastasis by interacting with METTL3. The PHGDH-METTL3 axis may serve as a potential clinical therapeutic target, offering new insights into the multifaceted roles of PHGDH in HCC metastasis.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2427-2438"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142856165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IRE1α inhibitor reduces cisplatin resistance in ovarian cancer by modulating IRE1α/XBP1 pathway. IRE1α抑制剂通过调节IRE1α/XBP1通路降低卵巢癌的顺铂耐药性
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-12-01 Epub Date: 2024-11-20 DOI: 10.1007/s13402-024-01010-z
Shiyi Lv, Lin Zhang, Min Wu, Shuangshuang Zhu, Yixue Wang, Layang Liu, Yunxuan Li, Ting Zhang, Yujie Wu, Huang Chen, Mingyao Liu, Zhengfang Yi

Ovarian cancer, a leading cause of gynecological cancer deaths globally, poses significant treatment challenges. Cisplatin (CDDP) is the first treatment choice for ovarian cancer and it is initially effective. However, 80% of ovarian cancer patients eventually relapse and develop resistance, resulting in chemotherapy failure. Therefore, finding new treatment combinations to overcome ovarian cancer resistance can provide a new tactic to improve the ovarian cancer patients' survival rate. We first identified activation of the Unfolded Protein Response (UPR) in CDDP-resistant ovarian cancer cells, implicating the IRE1α/XBP1 pathway in promoting resistance. Our findings demonstrate that inhibiting IRE1α signaling can re-sensitizes resistant cells to CDDP in vivo and in vitro, suggesting that IRE1α inhibitor used in conjunction with CDDP presumably could merge as a novel therapeutic strategy. Here, our research highlights the critical role of IRE1α signaling in mediating CDDP resistance, and paves the way for improved treatment options through combinatorial therapy.

卵巢癌是全球妇科癌症死亡的主要原因,给治疗带来了巨大挑战。顺铂(CDDP)是卵巢癌的首选治疗药物,而且在初期疗效显著。然而,80% 的卵巢癌患者最终会复发并产生抗药性,导致化疗失败。因此,寻找克服卵巢癌耐药性的新的治疗组合为提高卵巢癌患者的生存率提供了新的策略。我们首次发现 CDDP 耐药卵巢癌细胞中存在未折叠蛋白反应(UPR)的激活,这与促进耐药性的 IRE1α/XBP1 通路有关。我们的研究结果表明,抑制IRE1α信号传导可使体内和体外对CDDP耐药的细胞重新敏感,这表明IRE1α抑制剂与CDDP结合使用可能会成为一种新的治疗策略。在此,我们的研究强调了IRE1α信号传导在介导CDDP耐药性中的关键作用,并为通过组合疗法改善治疗方案铺平了道路。
{"title":"IRE1α inhibitor reduces cisplatin resistance in ovarian cancer by modulating IRE1α/XBP1 pathway.","authors":"Shiyi Lv, Lin Zhang, Min Wu, Shuangshuang Zhu, Yixue Wang, Layang Liu, Yunxuan Li, Ting Zhang, Yujie Wu, Huang Chen, Mingyao Liu, Zhengfang Yi","doi":"10.1007/s13402-024-01010-z","DOIUrl":"10.1007/s13402-024-01010-z","url":null,"abstract":"<p><p>Ovarian cancer, a leading cause of gynecological cancer deaths globally, poses significant treatment challenges. Cisplatin (CDDP) is the first treatment choice for ovarian cancer and it is initially effective. However, 80% of ovarian cancer patients eventually relapse and develop resistance, resulting in chemotherapy failure. Therefore, finding new treatment combinations to overcome ovarian cancer resistance can provide a new tactic to improve the ovarian cancer patients' survival rate. We first identified activation of the Unfolded Protein Response (UPR) in CDDP-resistant ovarian cancer cells, implicating the IRE1α/XBP1 pathway in promoting resistance. Our findings demonstrate that inhibiting IRE1α signaling can re-sensitizes resistant cells to CDDP in vivo and in vitro, suggesting that IRE1α inhibitor used in conjunction with CDDP presumably could merge as a novel therapeutic strategy. Here, our research highlights the critical role of IRE1α signaling in mediating CDDP resistance, and paves the way for improved treatment options through combinatorial therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2233-2246"},"PeriodicalIF":4.9,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142677501","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","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
Cellular Oncology
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: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