首页 > 最新文献

Cancer Cell International最新文献

英文 中文
CircUBA2 promotes the cancer stem cell-like properties of gastric cancer through upregulating STC1 via sponging miR-144-5p. CircUBA2通过海绵状miR-144-5p上调STC1,从而促进胃癌的癌症干细胞样特性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s12935-024-03423-0
Jia-Bin Wang, Tong-Xing Lin, Deng-Hui Fan, You-Xin Gao, Yu-Jing Chen, Yu-Kai Wu, Kai-Xiang Xu, Qing-Zhu Qiu, Ping Li, Jian-Wei Xie, Jian-Xian Lin, Qi-Yue Chen, Long-Long Cao, Chang-Ming Huang, Chao-Hui Zheng

Background: Cancer stem cells (CSCs) are critical factors that limit the effectiveness of gastric cancer (GC) therapy. Circular RNAs (circRNAs) are confirmed as important regulators of many cancers. However, their role in regulating CSC-like properties of GC remains largely unknown. Our study aimed to investigate the role of circUBA2 in CSC maintenance and the underlying mechanisms.

Methods: We identified circUBA2 as an upregulated gene using circRNA microarray analysis. qRT-PCR was used to examine the circUBA2 levels in normal and GC tissues. In vitro and in vivo functional assays were performed to validate the role of circUBA2 in proliferation, migration, metastasis and CSC-like properties of GC cell. The relationship between circUBA2, miR-144-5p and STC1 was characterised using bioinformatics analysis, a dual fluorescence reporter system, FISH, and RIP assays.

Results: CircUBA2 expression was significantly increased in GC tissues, and patients with GC with high circUBA2 expression had a poor prognosis. CircUBA2 enhances CSC-like properties of GC, thereby promoting cell proliferation, migration, and metastasis. Mechanistically, circUBA2 promoted GC malignancy and CSC-like properties by acting as a sponge for miR-144-5p to upregulate STC1 expression and further activate the IL-6/JAK2/STAT3 signaling pathway. More importantly, the ability of circUBA2 to enhance CSC-like properties was inhibited by tocilizumab, a humanised Interleukin-6 receptor (IL-6R) antibody. Thus, circUBA2 knockdown and tocilizumab synergistically inhibited CSC-like properties.

Conclusions: Our study demonstrated the critical role of circUBA2 in regulating CSC-like properties in GC. CircUBA2 may be a promising prognostic biomarker for GC.

背景:癌症干细胞(CSCs)是限制胃癌(GC)治疗效果的关键因素。环状 RNA(circRNA)已被证实是许多癌症的重要调控因子。然而,它们在调控胃癌 CSC 类特性中的作用在很大程度上仍不为人所知。我们的研究旨在探讨 circUBA2 在 CSC 维持中的作用及其潜在机制:方法:我们利用 circRNA 微阵列分析确定了 circUBA2 是一个上调基因。体外和体内功能测试验证了circUBA2在GC细胞增殖、迁移、转移和CSC样特性中的作用。通过生物信息学分析、双荧光报告系统、FISH和RIP检测,研究了circUBA2、miR-144-5p和STC1之间的关系:结果:CircUBA2在GC组织中的表达明显增加,高表达circUBA2的GC患者预后较差。CircUBA2增强了GC的CSC样特性,从而促进细胞增殖、迁移和转移。从机理上讲,circUBA2通过充当miR-144-5p的海绵来上调STC1的表达,并进一步激活IL-6/JAK2/STAT3信号通路,从而促进了GC的恶性程度和CSC样特性。更重要的是,人源化白细胞介素-6受体(IL-6R)抗体托西珠单抗抑制了circUBA2增强CSC样特性的能力。因此,circUBA2敲除和托珠单抗协同抑制了CSC样特性:我们的研究证明了circUBA2在调节GC中CSC样特性中的关键作用。结论:我们的研究证明了circUBA2在调节GC中CSC样特性中的关键作用,circUBA2可能是一种有前景的GC预后生物标志物。
{"title":"CircUBA2 promotes the cancer stem cell-like properties of gastric cancer through upregulating STC1 via sponging miR-144-5p.","authors":"Jia-Bin Wang, Tong-Xing Lin, Deng-Hui Fan, You-Xin Gao, Yu-Jing Chen, Yu-Kai Wu, Kai-Xiang Xu, Qing-Zhu Qiu, Ping Li, Jian-Wei Xie, Jian-Xian Lin, Qi-Yue Chen, Long-Long Cao, Chang-Ming Huang, Chao-Hui Zheng","doi":"10.1186/s12935-024-03423-0","DOIUrl":"10.1186/s12935-024-03423-0","url":null,"abstract":"<p><strong>Background: </strong>Cancer stem cells (CSCs) are critical factors that limit the effectiveness of gastric cancer (GC) therapy. Circular RNAs (circRNAs) are confirmed as important regulators of many cancers. However, their role in regulating CSC-like properties of GC remains largely unknown. Our study aimed to investigate the role of circUBA2 in CSC maintenance and the underlying mechanisms.</p><p><strong>Methods: </strong>We identified circUBA2 as an upregulated gene using circRNA microarray analysis. qRT-PCR was used to examine the circUBA2 levels in normal and GC tissues. In vitro and in vivo functional assays were performed to validate the role of circUBA2 in proliferation, migration, metastasis and CSC-like properties of GC cell. The relationship between circUBA2, miR-144-5p and STC1 was characterised using bioinformatics analysis, a dual fluorescence reporter system, FISH, and RIP assays.</p><p><strong>Results: </strong>CircUBA2 expression was significantly increased in GC tissues, and patients with GC with high circUBA2 expression had a poor prognosis. CircUBA2 enhances CSC-like properties of GC, thereby promoting cell proliferation, migration, and metastasis. Mechanistically, circUBA2 promoted GC malignancy and CSC-like properties by acting as a sponge for miR-144-5p to upregulate STC1 expression and further activate the IL-6/JAK2/STAT3 signaling pathway. More importantly, the ability of circUBA2 to enhance CSC-like properties was inhibited by tocilizumab, a humanised Interleukin-6 receptor (IL-6R) antibody. Thus, circUBA2 knockdown and tocilizumab synergistically inhibited CSC-like properties.</p><p><strong>Conclusions: </strong>Our study demonstrated the critical role of circUBA2 in regulating CSC-like properties in GC. CircUBA2 may be a promising prognostic biomarker for GC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302268/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multiple mechanisms contribute to acquired TRAIL resistance in multiple myeloma. 多种机制导致多发性骨髓瘤获得性TRAIL耐药。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s12935-024-03466-3
Fany V Ticona-Pérez, Xi Chen, Atanasio Pandiella, Elena Díaz-Rodríguez

Multiple Myeloma (MM) prognosis has recently improved thanks to the incorporation of new therapies to the clinic. Nonetheless, it is still a non-curable malignancy. Targeting cancer cells with agents inducing cell death has been an appealing alternative investigated over the years, as is the case of TRAIL, an agonist of DR4 and DR5 death receptors. This pathway, involved in apoptosis triggering, has demonstrated efficacy on MM cells. In this research, we have investigated the sensitivity of a panel of MM cells to this agent and generated TRAIL-resistant models by continuous culture of sensitive cells with this peptide. Using genomic and biochemical approaches, the mechanisms underlying resistance were investigated. In TRAIL-resistant cells, a strong reduction in cell-surface receptor levels was detected and impaired the apoptotic machinery to respond to the treatment, enabling cells to efficiently form the Death Inducing Signalling Complex. In addition, an upregulation of the inhibitory protein c-FLIP was detected. Even though the manipulation of these proteins was able to modify cellular responses to TRAIL, it was not complete, pointing to other mechanisms involved in TRAIL resistance.

由于临床上采用了新疗法,多发性骨髓瘤(MM)的预后最近有所改善。尽管如此,多发性骨髓瘤仍然是一种无法治愈的恶性肿瘤。使用诱导细胞死亡的药物靶向癌细胞是多年来研究的一种有吸引力的替代疗法,DR4 和 DR5 死亡受体的激动剂 TRAIL 就是一个例子。TRAIL 是一种 DR4 和 DR5 死亡受体激动剂,它参与触发细胞凋亡,已被证实对 MM 细胞有效。在这项研究中,我们调查了一组 MM 细胞对这种药物的敏感性,并通过用这种肽对敏感细胞进行连续培养,生成了 TRAIL 抗性模型。通过基因组学和生物化学方法,我们研究了产生耐药性的机制。在 TRAIL 抗性细胞中,检测到细胞表面受体水平大幅降低,从而影响了凋亡机制对治疗的反应,使细胞不能有效地形成死亡诱导信号复合物。此外,还检测到抑制蛋白 c-FLIP 的上调。尽管操纵这些蛋白能够改变细胞对 TRAIL 的反应,但这并不完全,这表明 TRAIL 抗性还涉及其他机制。
{"title":"Multiple mechanisms contribute to acquired TRAIL resistance in multiple myeloma.","authors":"Fany V Ticona-Pérez, Xi Chen, Atanasio Pandiella, Elena Díaz-Rodríguez","doi":"10.1186/s12935-024-03466-3","DOIUrl":"10.1186/s12935-024-03466-3","url":null,"abstract":"<p><p>Multiple Myeloma (MM) prognosis has recently improved thanks to the incorporation of new therapies to the clinic. Nonetheless, it is still a non-curable malignancy. Targeting cancer cells with agents inducing cell death has been an appealing alternative investigated over the years, as is the case of TRAIL, an agonist of DR4 and DR5 death receptors. This pathway, involved in apoptosis triggering, has demonstrated efficacy on MM cells. In this research, we have investigated the sensitivity of a panel of MM cells to this agent and generated TRAIL-resistant models by continuous culture of sensitive cells with this peptide. Using genomic and biochemical approaches, the mechanisms underlying resistance were investigated. In TRAIL-resistant cells, a strong reduction in cell-surface receptor levels was detected and impaired the apoptotic machinery to respond to the treatment, enabling cells to efficiently form the Death Inducing Signalling Complex. In addition, an upregulation of the inhibitory protein c-FLIP was detected. Even though the manipulation of these proteins was able to modify cellular responses to TRAIL, it was not complete, pointing to other mechanisms involved in TRAIL resistance.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141888539","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DDX19A promotes gastric cancer cell proliferation and migration by activating the PI3K/AKT pathway DDX19A 通过激活 PI3K/AKT 通路促进胃癌细胞增殖和迁移
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-03 DOI: 10.1186/s12935-024-03448-5
Yu Cheng, Yanjie Lu, Jing Xue, Xuemei Wang, Lili Zhou, Yu Luo, Yuhong Li
DEAD-box RNA helicase 19 A (DDX19A) is overexpressed in cervical squamous cell carcinoma. However, its role in gastric cancer remains unclear. The present study aimed to explore the role and underlying mechanism of DDX19A in the development of gastric cancer. The expression of DDX19A in gastric cancer and paracancerous tissues was evaluated through quantitative polymerase chain reaction, western blotting, and immunohistochemical staining. The biological functions of DDX19A in gastric cancer were determined using CCK8, plate colony-forming, and Transwell migration assays. The specific mechanism of DDX19A in gastric cancer cells was studied using western blotting, RNA-binding protein immunoprecipitation, mRNA half-life detection, and nuclear and cytoplasmic RNA isolation. DDX19A was highly expressed in gastric cancer and positively associated with malignant clinicopathological features and poor prognosis. Additionally, DDX19A promoted gastric cancer cell proliferation, migration, and epithelial–mesenchymal transition phenotypes. Mechanistically, DDX19A activated the PI3K/AKT pathway by upregulating phosphatidylinositol-3-kinase (PIK3CA) expression. Furthermore, DDX19A interacted with PIK3CA mRNA, stabilized it, and facilitated its export from the nucleus. Our study reveals a novel mechanism whereby DDX19A promotes the proliferation and migration of gastric cancer cells by enhancing the stability and nuclear export of PIK3CA mRNA, thereby activating the PI3K/AKT pathway.
DEAD-box RNA 螺旋酶 19 A(DDX19A)在宫颈鳞状细胞癌中过度表达。然而,它在胃癌中的作用仍不清楚。本研究旨在探讨DDX19A在胃癌发生中的作用及其内在机制。通过定量聚合酶链式反应、Western 印迹和免疫组化染色评估了 DDX19A 在胃癌和癌旁组织中的表达。通过CCK8、平板集落形成和Transwell迁移试验确定了DDX19A在胃癌中的生物学功能。利用 Western 印迹、RNA 结合蛋白免疫沉淀、mRNA 半衰期检测、核和细胞质 RNA 分离等方法研究了 DDX19A 在胃癌细胞中的特异性机制。DDX19A在胃癌中高表达,与恶性临床病理特征和不良预后呈正相关。此外,DDX19A还能促进胃癌细胞的增殖、迁移和上皮-间质转化表型。从机理上讲,DDX19A通过上调磷脂酰肌醇-3-激酶(PIK3CA)的表达激活了PI3K/AKT通路。此外,DDX19A 与 PIK3CA mRNA 相互作用,使其稳定,并促进其从细胞核中输出。我们的研究揭示了一种新的机制,即DDX19A通过增强PIK3CA mRNA的稳定性和核输出,从而激活PI3K/AKT通路,促进胃癌细胞的增殖和迁移。
{"title":"DDX19A promotes gastric cancer cell proliferation and migration by activating the PI3K/AKT pathway","authors":"Yu Cheng, Yanjie Lu, Jing Xue, Xuemei Wang, Lili Zhou, Yu Luo, Yuhong Li","doi":"10.1186/s12935-024-03448-5","DOIUrl":"https://doi.org/10.1186/s12935-024-03448-5","url":null,"abstract":"DEAD-box RNA helicase 19 A (DDX19A) is overexpressed in cervical squamous cell carcinoma. However, its role in gastric cancer remains unclear. The present study aimed to explore the role and underlying mechanism of DDX19A in the development of gastric cancer. The expression of DDX19A in gastric cancer and paracancerous tissues was evaluated through quantitative polymerase chain reaction, western blotting, and immunohistochemical staining. The biological functions of DDX19A in gastric cancer were determined using CCK8, plate colony-forming, and Transwell migration assays. The specific mechanism of DDX19A in gastric cancer cells was studied using western blotting, RNA-binding protein immunoprecipitation, mRNA half-life detection, and nuclear and cytoplasmic RNA isolation. DDX19A was highly expressed in gastric cancer and positively associated with malignant clinicopathological features and poor prognosis. Additionally, DDX19A promoted gastric cancer cell proliferation, migration, and epithelial–mesenchymal transition phenotypes. Mechanistically, DDX19A activated the PI3K/AKT pathway by upregulating phosphatidylinositol-3-kinase (PIK3CA) expression. Furthermore, DDX19A interacted with PIK3CA mRNA, stabilized it, and facilitated its export from the nucleus. Our study reveals a novel mechanism whereby DDX19A promotes the proliferation and migration of gastric cancer cells by enhancing the stability and nuclear export of PIK3CA mRNA, thereby activating the PI3K/AKT pathway.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141886185","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
MSI2 regulates NLK-mediated EMT and PI3K/AKT/mTOR pathway to promote pancreatic cancer progression MSI2 调控 NLK 介导的 EMT 和 PI3K/AKT/mTOR 通路,促进胰腺癌进展
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-03 DOI: 10.1186/s12935-024-03444-9
Longping Huang, Jian Sun, Yuteng Ma, He Chen, Chen Tian, Ming Dong
The incidence of pancreatic cancer is increasing by years, and the 5-year survival rate is very low. Our team have revealed that Musashi2 (MSI2) could promote aggressive behaviors in pancreatic cancer by downregulating Numb and p53. MSI2 also facilitates EMT in pancreatic cancer induced by EGF through the ZEB1-ERK/MAPK signaling pathway. This study aims to further explore the molecular mechanisms of MSI2-regulated downstream pathways in pancreatic cancer. In vitro and in vivo experiments were conducted to investigate the role and mechanism of MSI2 in promoting malignant behaviors of pancreatic cancer through regulation of NLK. Genes closely related to MSI2 were screened from the GEPIA and TCGA databases. We found that NLK showed the most significant changes in mRNA levels with consistent changes following MSI2 interference and overexpression. The high correlation between MSI2 and NLK was also observed at the protein level. Multivariate analysis revealed that both MSI2 and NLK were independent adverse indicators of survival in pancreatic cancer patients, as well as join together. In vitro, silencing or overexpressing NLK altered cell invasion and migration, by regulating EMT and the PI3K-AKT-mTOR pathway. Silencing MSI2 reduced protein expression in the EMT and PI3K-AKT-mTOR pathways, leading to decreased cell invasion and migration abilities, while these effects could be reversed by overexpression of NLK. In vivo, MSI2 silencing inhibited liver metastasis, which could be reversed by overexpressing NLK. Mechanistically, MSI2 directly binds to the translation regulatory region of NLK mRNA at positions 79–87 nt, enhancing its transcriptional activity and exerting post-transcriptional regulatory roles. The analysis of molecular docking showed the close relationship between MSI2 and NLK in pancreatic cancer patients. Our findings elucidate the regulatory mechanisms of the MSI2-NLK axis in modulating aggressive behaviors of pancreatic cancer cells, which providing new evidence for therapeutic strategies in pancreatic cancer.
胰腺癌的发病率逐年上升,5 年生存率非常低。我们的团队发现,Musashi2(MSI2)可通过下调Numb和p53促进胰腺癌的侵袭行为。MSI2 还可通过 ZEB1-ERK/MAPK 信号通路促进 EGF 诱导的胰腺癌 EMT。本研究旨在进一步探讨MSI2调控胰腺癌下游通路的分子机制。通过体外和体内实验,研究MSI2通过调控NLK促进胰腺癌恶性行为的作用和机制。我们从GEPIA和TCGA数据库中筛选了与MSI2密切相关的基因。我们发现,NLK在MSI2干扰和过表达后的mRNA水平变化最为显著,且变化一致。在蛋白质水平上也观察到了MSI2和NLK之间的高度相关性。多变量分析显示,MSI2和NLK是胰腺癌患者生存的独立不利指标,同时也是共同的不利指标。在体外,沉默或过表达NLK可通过调节EMT和PI3K-AKT-mTOR通路改变细胞的侵袭和迁移。沉默MSI2会降低EMT和PI3K-AKT-mTOR通路的蛋白表达,导致细胞侵袭和迁移能力下降,而过表达NLK则可逆转这些影响。在体内,沉默MSI2可抑制肝转移,而过表达NLK则可逆转这种效应。从机制上看,MSI2直接与NLK mRNA翻译调控区79-87 nt位结合,增强其转录活性并发挥转录后调控作用。分子对接分析表明,在胰腺癌患者中,MSI2与NLK关系密切。我们的研究结果阐明了MSI2-NLK轴在调节胰腺癌细胞侵袭行为中的调控机制,为胰腺癌的治疗策略提供了新的证据。
{"title":"MSI2 regulates NLK-mediated EMT and PI3K/AKT/mTOR pathway to promote pancreatic cancer progression","authors":"Longping Huang, Jian Sun, Yuteng Ma, He Chen, Chen Tian, Ming Dong","doi":"10.1186/s12935-024-03444-9","DOIUrl":"https://doi.org/10.1186/s12935-024-03444-9","url":null,"abstract":"The incidence of pancreatic cancer is increasing by years, and the 5-year survival rate is very low. Our team have revealed that Musashi2 (MSI2) could promote aggressive behaviors in pancreatic cancer by downregulating Numb and p53. MSI2 also facilitates EMT in pancreatic cancer induced by EGF through the ZEB1-ERK/MAPK signaling pathway. This study aims to further explore the molecular mechanisms of MSI2-regulated downstream pathways in pancreatic cancer. In vitro and in vivo experiments were conducted to investigate the role and mechanism of MSI2 in promoting malignant behaviors of pancreatic cancer through regulation of NLK. Genes closely related to MSI2 were screened from the GEPIA and TCGA databases. We found that NLK showed the most significant changes in mRNA levels with consistent changes following MSI2 interference and overexpression. The high correlation between MSI2 and NLK was also observed at the protein level. Multivariate analysis revealed that both MSI2 and NLK were independent adverse indicators of survival in pancreatic cancer patients, as well as join together. In vitro, silencing or overexpressing NLK altered cell invasion and migration, by regulating EMT and the PI3K-AKT-mTOR pathway. Silencing MSI2 reduced protein expression in the EMT and PI3K-AKT-mTOR pathways, leading to decreased cell invasion and migration abilities, while these effects could be reversed by overexpression of NLK. In vivo, MSI2 silencing inhibited liver metastasis, which could be reversed by overexpressing NLK. Mechanistically, MSI2 directly binds to the translation regulatory region of NLK mRNA at positions 79–87 nt, enhancing its transcriptional activity and exerting post-transcriptional regulatory roles. The analysis of molecular docking showed the close relationship between MSI2 and NLK in pancreatic cancer patients. Our findings elucidate the regulatory mechanisms of the MSI2-NLK axis in modulating aggressive behaviors of pancreatic cancer cells, which providing new evidence for therapeutic strategies in pancreatic cancer.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141886182","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
Correction: MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome 更正:以mTOR为靶点的微RNA是改善放疗效果的治疗药物
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-03 DOI: 10.1186/s12935-024-03461-8
Shahram Taeb, Davoud Rostamzadeh, Seyed Mohammad Amini, Mohammad Rahmati, Mohammad Eftekhari, Arash Safari, Masoud Najafi

Correction to: Cancer Cell International (2024) 24:233

https://doi.org/10.1186/s12935-024-03420-3

In this article [1], the affiliation details for the author Arash Safari were incorrectly given as ‘Department of Radiology, Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz 71439 − 14693, Iran’ but should have been ‘Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran and Department of Radiology, Paramedical School, Shiraz University of Medical Sciences, Shiraz, Iran’.

  1. Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Eftekhari M, Safari A, Najafi M. MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome. Cancer Cell Int. 2024;24(1):233.

    Article PubMed PubMed Central Google Scholar

Download references

Authors and Affiliations

  1. Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran

    Shahram Taeb

  2. Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA

    Davoud Rostamzadeh

  3. Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran

    Seyed Mohammad Amini

  4. Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran

    Mohammad Rahmati & Mohammad Eftekhari

  5. Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran

    Arash Safari

  6. Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran

    Arash Safari

  7. Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran

    Masoud Najafi

  8. Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran

    Masoud Najafi

  9. Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran

    Masoud Najafi

Authors
  1. Shahram TaebView author publications

    You can also search for this author in PubMed Google Scholar

  2. Davoud RostamzadehView author publications

    You can also search for this author in

Correction to:Cancer Cell International (2024) 24:233https://doi.org/10.1186/s12935-024-03420-3 在这篇文章[1]中,作者Arash Safari的单位信息错误地填写为 "Department of Radiology, Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences、Shiraz 71439 - 14693, Iran"(伊朗设拉子 71439 - 14693),但应该是 "Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran and Department of Radiology, Paramedical School, Shiraz University of Medical Sciences, Shiraz, Iran"。Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Eftekhari M, Safari A, Najafi M. MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome.Cancer Cell Int.Article PubMed PubMed Central Google Scholar Download references作者及工作单位伊朗拉什特吉兰医科大学辅助医疗科学学院放射学系Shahram Taeb康涅狄格大学健康中心免疫学系、Seyed Mohammad AminiDepartment of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, IranMohammad Rahmati &;Mohammad EftekhariIonizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, IranArash SafariDepartment of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, IranArash SafariRadiology and Nuclear Medicine Department、伊朗克尔曼沙赫,克尔曼沙赫医科大学辅助医疗科学学院放射学与核医学系伊朗克尔曼沙赫,克尔曼沙赫医科大学健康技术研究所医学生物学研究中心伊朗克尔曼沙赫,克尔曼沙赫医科大学健康技术研究所医学技术研究中心、伊朗Masoud Najafi作者Shahram Taeb查看作者发表的论文您也可以在PubMed Google ScholarDavoud Rostamzadeh查看作者发表的论文您也可以在PubMed Google ScholarSeyed Mohammad Amini查看作者发表的论文您也可以在PubMed Google ScholarMohammad Rahmati查看作者发表的论文您也可以在PubMed Google ScholarSeyed Mohammad Amini查看作者发表的论文您也可以在PubMed Google ScholarMohammad Rahmati查看作者发表的论文您也可以在PubMed Google Scholar搜索该作者Mohammad Eftekhari查看作者发表的作品您也可以在 PubMed Google ScholarArash Safari查看作者发表的作品您也可以在 PubMed Google ScholarMasoud Najafi查看作者发表的作品您也可以在 PubMed Google ScholarCorresponding author给 Masoud Najafi 的回信。出版商注释Springer Nature对出版地图中的管辖权主张和机构隶属关系保持中立。原文的在线版本可在以下网址找到:https://doi.org/10.1186/s12935-024-03420-3.Open Access 本文采用知识共享署名 4.0 国际许可协议进行许可,该协议允许以任何媒介或格式使用、共享、改编、分发和复制,只要您适当注明原作者和来源,提供知识共享许可协议的链接,并说明是否进行了修改。本文中的图片或其他第三方材料均包含在文章的知识共享许可协议中,除非在材料的署名栏中另有说明。如果材料未包含在文章的知识共享许可协议中,且您打算使用的材料不符合法律规定或超出许可使用范围,则您需要直接从版权所有者处获得许可。要查看该许可的副本,请访问 http://creativecommons.org/licenses/by/4.0/。除非在数据的信用行中另有说明,否则创作共用公共领域专用免责声明 (http://creativecommons.org/publicdomain/zero/1.0/) 适用于本文提供的数据。转载与许可引用本文Taeb, S., Rostamzadeh, D., Amini, S.M. et al. Correction:以 mTOR 为靶点的 MicroRNAs 可改善放疗效果。Cancer Cell Int 24, 274 (2024). https://doi.org/10.1186/s12935-024-03461-8Download citationAccepted: 26 July 2024Published: 03 August 2024DOI: https://doi.org/10.1186/s12935-024-03461-8Share this articleAnyone you share the following link with will be able to read this content:Get shareable linkSorry, a shareable link is not currently available for this article.Copy to clipboard Provided by th
{"title":"Correction: MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome","authors":"Shahram Taeb, Davoud Rostamzadeh, Seyed Mohammad Amini, Mohammad Rahmati, Mohammad Eftekhari, Arash Safari, Masoud Najafi","doi":"10.1186/s12935-024-03461-8","DOIUrl":"https://doi.org/10.1186/s12935-024-03461-8","url":null,"abstract":"<p><b>Correction to: Cancer Cell International (2024) 24:233</b></p><p><b>https://doi.org/10.1186/s12935-024-03420-3</b></p><p> In this article [1], the affiliation details for the author Arash Safari were incorrectly given as ‘Department of Radiology, Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz 71439 − 14693, Iran’ but should have been ‘Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran and Department of Radiology, Paramedical School, Shiraz University of Medical Sciences, Shiraz, Iran’.</p><ol data-track-component=\"outbound reference\" data-track-context=\"references section\"><li data-counter=\"1.\"><p>Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Eftekhari M, Safari A, Najafi M. MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome. Cancer Cell Int. 2024;24(1):233.</p><p>Article PubMed PubMed Central Google Scholar </p></li></ol><p>Download references<svg aria-hidden=\"true\" focusable=\"false\" height=\"16\" role=\"img\" width=\"16\"><use xlink:href=\"#icon-eds-i-download-medium\" xmlns:xlink=\"http://www.w3.org/1999/xlink\"></use></svg></p><h3>Authors and Affiliations</h3><ol><li><p>Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran</p><p>Shahram Taeb</p></li><li><p>Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA</p><p>Davoud Rostamzadeh</p></li><li><p>Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran</p><p>Seyed Mohammad Amini</p></li><li><p>Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran</p><p>Mohammad Rahmati &amp; Mohammad Eftekhari</p></li><li><p>Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran</p><p>Arash Safari</p></li><li><p>Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran</p><p>Arash Safari</p></li><li><p>Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran</p><p>Masoud Najafi</p></li><li><p>Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran</p><p>Masoud Najafi</p></li><li><p>Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran</p><p>Masoud Najafi</p></li></ol><span>Authors</span><ol><li><span>Shahram Taeb</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Davoud Rostamzadeh</span>View author publications<p>You can also search for this author in <s","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141886183","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
TERT upstream promoter methylation regulates TERT expression and acts as a therapeutic target in TERT promoter mutation-negative thyroid cancer TERT 上游启动子甲基化调控 TERT 的表达,并成为 TERT 启动子突变阴性甲状腺癌的治疗靶点
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-03 DOI: 10.1186/s12935-024-03459-2
Shiyong Li, Guanghui Hu, Yulu Chen, Ye Sang, Qin Tang, Rengyun Liu
DNA hypermethylation and hotspot mutations were frequently observed in the upstream and core promoter of telomerase reverse transcriptase (TERT), respectively, and they were associated with increased TERT expression and adverse clinical outcomes in thyroid cancer. In TERT promoter mutant cancer cells, the hypomethylated TERT mutant allele was active and the hypermethylated TERT wild-type allele was silenced. However, whether and how the upstream promoter methylation regulates TERT expression in TERT mutation-negative cells were largely unknown. DNA demethylating agents 5-azacytidine and decitabine and a genomic locus-specific demethylation system based on dCas9-TET1 were used to assess the effects of TERT upstream promoter methylation on TERT expression, cell growth and apoptosis of thyroid cancer cells. Regulatory proteins binding to TERT promoter were identified by CRISPR affinity purification in situ of regulatory elements (CAPTURE) combined with mass spectrometry. The enrichments of selected regulatory proteins and histone modifications were evaluated by chromatin immunoprecipitation. The level of DNA methylation at TERT upstream promoter and expression of TERT were significantly decreased after treatment with 5-azacytidine or decitabine in TERT promoter wild-type thyroid cancer cells. Genomic locus-specific demethylation of TERT upstream promoter induced TERT downregulation, along with cell apoptosis and growth inhibition. Consistently, demethylating agents sharply inhibited the growth of thyroid cancer cells harboring hypermethylated TERT but had little effect on cells with TERT hypomethylation. Moreover, we identified that the chromatin remodeling protein CHD4 binds to methylated TERT upstream promoter and promotes its transcription by suppressing the enrichment of H3K9me3 and H3K27me3 at TERT promoter. This study uncovered the mechanism of promoter methylation mediated TERT activation in TERT promoter mutation-negative thyroid cancer cells and indicated TERT upstream promoter methylation as a therapeutic target for thyroid cancer.
在端粒酶逆转录酶(TERT)的上游启动子和核心启动子中经常分别观察到DNA高甲基化和热点突变,它们与TERT表达的增加和甲状腺癌的不良临床结果有关。在TERT启动子突变的癌细胞中,低甲基化的TERT突变等位基因是活跃的,而高甲基化的TERT野生型等位基因是沉默的。然而,上游启动子甲基化是否以及如何调控TERT突变阴性细胞中的TERT表达,在很大程度上还是未知数。研究人员利用DNA去甲基化药物5-氮杂胞苷和地西他滨以及基于dCas9-TET1的基因组位点特异性去甲基化系统,评估了TERT上游启动子甲基化对甲状腺癌细胞TERT表达、细胞生长和凋亡的影响。通过 CRISPR 亲和纯化原位调控元件(CAPTURE)并结合质谱分析,鉴定了与 TERT 启动子结合的调控蛋白。染色质免疫沉淀法评估了所选调控蛋白和组蛋白修饰的富集情况。用 5-azacytidine 或地西他滨处理 TERT 启动子野生型甲状腺癌细胞后,TERT 上游启动子的 DNA 甲基化水平和 TERT 的表达均显著下降。TERT上游启动子的基因组位点特异性去甲基化诱导TERT下调,同时导致细胞凋亡和生长抑制。同样,去甲基化药物能显著抑制TERT高甲基化的甲状腺癌细胞的生长,但对TERT低甲基化的细胞影响甚微。此外,我们还发现染色质重塑蛋白CHD4与甲基化的TERT上游启动子结合,并通过抑制TERT启动子上H3K9me3和H3K27me3的富集来促进其转录。该研究揭示了TERT启动子突变阴性甲状腺癌细胞中启动子甲基化介导的TERT激活机制,并指出TERT上游启动子甲基化是甲状腺癌的治疗靶点。
{"title":"TERT upstream promoter methylation regulates TERT expression and acts as a therapeutic target in TERT promoter mutation-negative thyroid cancer","authors":"Shiyong Li, Guanghui Hu, Yulu Chen, Ye Sang, Qin Tang, Rengyun Liu","doi":"10.1186/s12935-024-03459-2","DOIUrl":"https://doi.org/10.1186/s12935-024-03459-2","url":null,"abstract":"DNA hypermethylation and hotspot mutations were frequently observed in the upstream and core promoter of telomerase reverse transcriptase (TERT), respectively, and they were associated with increased TERT expression and adverse clinical outcomes in thyroid cancer. In TERT promoter mutant cancer cells, the hypomethylated TERT mutant allele was active and the hypermethylated TERT wild-type allele was silenced. However, whether and how the upstream promoter methylation regulates TERT expression in TERT mutation-negative cells were largely unknown. DNA demethylating agents 5-azacytidine and decitabine and a genomic locus-specific demethylation system based on dCas9-TET1 were used to assess the effects of TERT upstream promoter methylation on TERT expression, cell growth and apoptosis of thyroid cancer cells. Regulatory proteins binding to TERT promoter were identified by CRISPR affinity purification in situ of regulatory elements (CAPTURE) combined with mass spectrometry. The enrichments of selected regulatory proteins and histone modifications were evaluated by chromatin immunoprecipitation. The level of DNA methylation at TERT upstream promoter and expression of TERT were significantly decreased after treatment with 5-azacytidine or decitabine in TERT promoter wild-type thyroid cancer cells. Genomic locus-specific demethylation of TERT upstream promoter induced TERT downregulation, along with cell apoptosis and growth inhibition. Consistently, demethylating agents sharply inhibited the growth of thyroid cancer cells harboring hypermethylated TERT but had little effect on cells with TERT hypomethylation. Moreover, we identified that the chromatin remodeling protein CHD4 binds to methylated TERT upstream promoter and promotes its transcription by suppressing the enrichment of H3K9me3 and H3K27me3 at TERT promoter. This study uncovered the mechanism of promoter methylation mediated TERT activation in TERT promoter mutation-negative thyroid cancer cells and indicated TERT upstream promoter methylation as a therapeutic target for thyroid cancer.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141886184","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
ARAP1-AS1: a novel long non-coding RNA with a vital regulatory role in human cancer development ARAP1-AS1:在人类癌症发展过程中发挥重要调控作用的新型长非编码 RNA
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1186/s12935-024-03435-w
Jialing Wang, Hongliang Luo, Lu Yang, Huazhao Yuan
Long non-coding RNAs (lncRNAs) have garnered significant attention in biomedical research due to their pivotal roles in gene expression regulation and their association with various human diseases. Among these lncRNAs, ArfGAP With RhoGAP Domain, Ankyrin Repeat, And PH Domain 1 - Antisense RNA 1 (ARAP1-AS1) has recently emerged as an novel oncogenic player. ARAP1-AS1 is prominently overexpressed in numerous solid tumors and wields influence by modulating gene expression and signaling pathways. This regulatory impact is realized through dual mechanisms, involving both competitive interactions with microRNAs and direct protein binding. ARAP1-AS1 assumes an important role in driving tumorigenesis and malignant tumor progression, affecting biological characteristics such as tumor expansion and metastasis. This paper provides a concise review of the regulatory role of ARAP1-AS1 in malignant tumors and discuss its potential clinical applications as a biomarker and therapeutic target. We also address existing knowledge gaps and suggest avenues for future research. ARAP1-AS1 serves as a prototypical example within the burgeoning field of lncRNA studies, offering insights into the broader landscape of non-coding RNA molecules. This investigation enhances our comprehension of the complex mechanisms that govern the progression of cancer.
由于长非编码 RNA(lncRNA)在基因表达调控中的关键作用及其与各种人类疾病的关联,它们在生物医学研究中备受关注。在这些 lncRNA 中,ArfGAP With RhoGAP Domain, Ankyrin Repeat, And PH Domain 1 - Antisense RNA 1(ARAP1-AS1)最近成为一种新型致癌因子。ARAP1-AS1 在许多实体瘤中显著过表达,并通过调节基因表达和信号通路产生影响。这种调控影响是通过双重机制实现的,既涉及与 microRNA 的竞争性相互作用,也涉及与蛋白质的直接结合。ARAP1-AS1 在推动肿瘤发生和恶性肿瘤进展方面发挥着重要作用,影响着肿瘤扩张和转移等生物学特征。本文简要回顾了 ARAP1-AS1 在恶性肿瘤中的调控作用,并讨论了其作为生物标记物和治疗靶点的潜在临床应用。我们还探讨了现有的知识空白,并提出了未来的研究方向。ARAP1-AS1 是正在蓬勃发展的 lncRNA 研究领域中的一个典型例子,为我们了解非编码 RNA 分子的更广阔领域提供了见解。这项研究加深了我们对制约癌症进展的复杂机制的理解。
{"title":"ARAP1-AS1: a novel long non-coding RNA with a vital regulatory role in human cancer development","authors":"Jialing Wang, Hongliang Luo, Lu Yang, Huazhao Yuan","doi":"10.1186/s12935-024-03435-w","DOIUrl":"https://doi.org/10.1186/s12935-024-03435-w","url":null,"abstract":"Long non-coding RNAs (lncRNAs) have garnered significant attention in biomedical research due to their pivotal roles in gene expression regulation and their association with various human diseases. Among these lncRNAs, ArfGAP With RhoGAP Domain, Ankyrin Repeat, And PH Domain 1 - Antisense RNA 1 (ARAP1-AS1) has recently emerged as an novel oncogenic player. ARAP1-AS1 is prominently overexpressed in numerous solid tumors and wields influence by modulating gene expression and signaling pathways. This regulatory impact is realized through dual mechanisms, involving both competitive interactions with microRNAs and direct protein binding. ARAP1-AS1 assumes an important role in driving tumorigenesis and malignant tumor progression, affecting biological characteristics such as tumor expansion and metastasis. This paper provides a concise review of the regulatory role of ARAP1-AS1 in malignant tumors and discuss its potential clinical applications as a biomarker and therapeutic target. We also address existing knowledge gaps and suggest avenues for future research. ARAP1-AS1 serves as a prototypical example within the burgeoning field of lncRNA studies, offering insights into the broader landscape of non-coding RNA molecules. This investigation enhances our comprehension of the complex mechanisms that govern the progression of cancer.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141870255","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
Oncogenic LINC00698 suppresses apoptosis of melanoma stem cells to promote tumorigenesis via LINC00698-miR-3132-TCF7/hnRNPM axis 致癌基因 LINC00698 通过 LINC00698-miR-3132-TCF7/hnRNPM 轴抑制黑色素瘤干细胞凋亡以促进肿瘤发生
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-28 DOI: 10.1186/s12935-024-03408-z
Anas Mohammed, Ahmad Khan, Xiaobo Zhang
Melanoma progression depends on melanoma stem cells (MSCs), which are distinguished by the distinct dysregulated genes. As the key factors in the dysregulation of genes, long non-coding RNAs (lncRNAs) take great effects on MSCs. However, the underlying mechanism of lncRNAs in MSCs has not been extensively characterized. To address the roles of lncRNAs in MSCs, LINC00698 was characterized in this study. The results revealed that LINC00698 was upregulated in MSCs, showing its important role in MSCs. The further data indicated that the LINC00698 silencing triggered cell cycle arrest in the G0/G1 phase and apoptosis of MSCs. LINC00698 could directly interact with miR-3132 to upregulate the expression of TCF7, which was required for sustaining the stemness and the tumorigenic potency of MSCs. At the same time, LINC00698 could bind to the hnRNPM protein to enhance the protein stability, thus suppressing apoptosis and promoting the stemness of MSCs. Furthermore, the in vivo data demonstrated that LINC00698 was essential for tumorigenesis of MSCs via the LINC00698-miR-3132-TCF7/hnRNPM axis. Therefore, our findings contributed novel insights into the underlying mechanism of melanoma progression.
黑色素瘤的发展依赖于黑色素瘤干细胞(MSCs),而MSCs则以不同的失调基因为特征。作为基因失调的关键因素,长非编码 RNA(lncRNA)对间充质干细胞有很大影响。然而,lncRNAs在间充质干细胞中的内在机制尚未得到广泛表征。为了研究 lncRNA 在间充质干细胞中的作用,本研究对 LINC00698 进行了定性。结果显示,LINC00698 在间充质干细胞中上调,显示了其在间充质干细胞中的重要作用。进一步的数据表明,沉默 LINC00698 会导致间充质干细胞的细胞周期停滞在 G0/G1 期并发生凋亡。LINC00698可直接与miR-3132相互作用,上调TCF7的表达,而TCF7是维持间充质干细胞干性和致瘤性所必需的。同时,LINC00698能与hnRNPM蛋白结合,增强蛋白的稳定性,从而抑制间充质干细胞的凋亡并促进其干性。此外,体内数据表明,LINC00698通过LINC00698-miR-3132-TCF7/hnRNPM轴对间叶干细胞的肿瘤发生至关重要。因此,我们的研究结果为黑色素瘤进展的内在机制提供了新的见解。
{"title":"Oncogenic LINC00698 suppresses apoptosis of melanoma stem cells to promote tumorigenesis via LINC00698-miR-3132-TCF7/hnRNPM axis","authors":"Anas Mohammed, Ahmad Khan, Xiaobo Zhang","doi":"10.1186/s12935-024-03408-z","DOIUrl":"https://doi.org/10.1186/s12935-024-03408-z","url":null,"abstract":"Melanoma progression depends on melanoma stem cells (MSCs), which are distinguished by the distinct dysregulated genes. As the key factors in the dysregulation of genes, long non-coding RNAs (lncRNAs) take great effects on MSCs. However, the underlying mechanism of lncRNAs in MSCs has not been extensively characterized. To address the roles of lncRNAs in MSCs, LINC00698 was characterized in this study. The results revealed that LINC00698 was upregulated in MSCs, showing its important role in MSCs. The further data indicated that the LINC00698 silencing triggered cell cycle arrest in the G0/G1 phase and apoptosis of MSCs. LINC00698 could directly interact with miR-3132 to upregulate the expression of TCF7, which was required for sustaining the stemness and the tumorigenic potency of MSCs. At the same time, LINC00698 could bind to the hnRNPM protein to enhance the protein stability, thus suppressing apoptosis and promoting the stemness of MSCs. Furthermore, the in vivo data demonstrated that LINC00698 was essential for tumorigenesis of MSCs via the LINC00698-miR-3132-TCF7/hnRNPM axis. Therefore, our findings contributed novel insights into the underlying mechanism of melanoma progression.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141771112","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
Uncovering novel mechanisms of chitinase-3-like protein 1 in driving inflammation-associated cancers 揭示几丁质酶-3样蛋白1驱动炎症相关癌症的新机制
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-27 DOI: 10.1186/s12935-024-03425-y
Yan Fan, Yuan Meng, Xingwei Hu, Jianhua Liu, Xiaosong Qin
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that is induced and regulated by multiple factors during inflammation in enteritis, pneumonia, asthma, arthritis, and other diseases. It is associated with the deterioration of the inflammatory environment in tissues with chronic inflammation caused by microbial infection or autoimmune diseases. The expression of CHI3L1 expression is upregulated in several malignant tumors, underscoring the crucial role of chronic inflammation in the initiation and progression of cancer. While the precise mechanism connecting inflammation and cancer is unclear, the involvement of CHI3L1 is involved in chronic inflammation, suggesting its role as a contributing factor to in the link between inflammation and cancer. CHI3L1 can aggravate DNA oxidative damage, induce the cancerous phenotype, promote the development of a tumor inflammatory environment and angiogenesis, inhibit immune cells, and promote cancer cell growth, invasion, and migration. Furthermore, it participates in the initiation of cancer progression and metastasis by binding with transmembrane receptors to mediate intracellular signal transduction. Based on the current research on CHI3L1, we explore introduce the receptors that interact with CHI3L1 along with the signaling pathways that may be triggered during chronic inflammation to enhance tumorigenesis and progression. In the last section of the article, we provide a brief overview of anti-inflammatory therapies that target CHI3L1.
几丁质酶-3 样蛋白 1(CHI3L1)是一种分泌型糖蛋白,在肠炎、肺炎、哮喘、关节炎和其他疾病的炎症过程中受到多种因素的诱导和调节。它与微生物感染或自身免疫性疾病引起的慢性炎症组织中炎症环境的恶化有关。CHI3L1 的表达在多种恶性肿瘤中上调,凸显了慢性炎症在癌症发生和发展中的关键作用。虽然炎症与癌症之间的确切联系机制尚不清楚,但 CHI3L1 参与了慢性炎症,这表明它是炎症与癌症之间联系的一个促成因素。CHI3L1 可加重 DNA 氧化损伤,诱导癌症表型,促进肿瘤炎症环境的发展和血管生成,抑制免疫细胞,促进癌细胞生长、侵袭和迁移。此外,它还通过与跨膜受体结合,介导细胞内信号转导,从而参与癌症进展和转移的启动。基于目前对CHI3L1的研究,我们探讨介绍了与CHI3L1相互作用的受体,以及在慢性炎症过程中可能被触发以促进肿瘤发生和发展的信号通路。在文章的最后一部分,我们将简要介绍针对 CHI3L1 的抗炎疗法。
{"title":"Uncovering novel mechanisms of chitinase-3-like protein 1 in driving inflammation-associated cancers","authors":"Yan Fan, Yuan Meng, Xingwei Hu, Jianhua Liu, Xiaosong Qin","doi":"10.1186/s12935-024-03425-y","DOIUrl":"https://doi.org/10.1186/s12935-024-03425-y","url":null,"abstract":"Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that is induced and regulated by multiple factors during inflammation in enteritis, pneumonia, asthma, arthritis, and other diseases. It is associated with the deterioration of the inflammatory environment in tissues with chronic inflammation caused by microbial infection or autoimmune diseases. The expression of CHI3L1 expression is upregulated in several malignant tumors, underscoring the crucial role of chronic inflammation in the initiation and progression of cancer. While the precise mechanism connecting inflammation and cancer is unclear, the involvement of CHI3L1 is involved in chronic inflammation, suggesting its role as a contributing factor to in the link between inflammation and cancer. CHI3L1 can aggravate DNA oxidative damage, induce the cancerous phenotype, promote the development of a tumor inflammatory environment and angiogenesis, inhibit immune cells, and promote cancer cell growth, invasion, and migration. Furthermore, it participates in the initiation of cancer progression and metastasis by binding with transmembrane receptors to mediate intracellular signal transduction. Based on the current research on CHI3L1, we explore introduce the receptors that interact with CHI3L1 along with the signaling pathways that may be triggered during chronic inflammation to enhance tumorigenesis and progression. In the last section of the article, we provide a brief overview of anti-inflammatory therapies that target CHI3L1.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141771113","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
Integrative analysis of aging-related genes reveals CEBPA as a novel therapeutic target in non-small cell lung cancer 对衰老相关基因的整合分析发现 CEBPA 是治疗非小细胞肺癌的新靶点
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-27 DOI: 10.1186/s12935-024-03457-4
Jiaqi Zhu, Xiaoren Zhu, Conglin Shi, Qixuan Li, Yun Jiang, Xingyou Chen, Pingping Sun, Yi Jin, Tianyi Wang, Jianle Chen
To explore the impact of ARGs on the prognosis of NSCLC, and its correlation with clinicopathological parameters and immune microenvironment. Preliminary research on the biological functions of CEBPA in NSCLC. Using consensus clustering analysis to identify molecular subtypes of ARGs in NSCLC patients; employing LASSO regression and multivariate Cox analysis to select 7 prognostic risk genes and construct a prognostic risk model; validating independent prognostic factors of NSCLC using forest plot analysis; analyzing immune microenvironment correlations using ESTIMATE and ssGSEA; assessing correlations between prognostic risk genes via qPCR and Western blot in NSCLC; measuring mRNA and protein expression levels of knocked down and overexpressed CEBPA in NSCLC using CCK-8 and EdU assays; evaluating the effects of knocked down and overexpressed CEBPA on cell proliferation using Transwell experiments; examining the correlation of CEBPA with T cells and B cells using mIHC analysis. Consensus clustering analysis identified three molecular subtypes, suggesting significant differential expression of these ARGs in NSCLC prognosis and clinical pathological parameters. There was significant differential expression between the two risk groups in the prognostic risk model, with P < 0.001. The risk score of the prognostic risk model was also P < 0.001. CEBPA exhibited higher mRNA and protein expression levels in NSCLC cell lines. Knockdown of CEBPA significantly reduced mRNA and protein expression levels of CEBPB, YWHAZ, ABL1, and CDK1 in H1650 and A549 cells. siRNA-mediated knockdown of CEBPA markedly inhibited proliferation, migration, and invasion of NSCLC cells, whereas overexpression of CEBPA showed the opposite trend. mIHC results indicated a significant increase in CD3 + CD4+, CD3 + CD8+, and CD20 + cell counts in the high CEBPA expression group. The risk score of the prognostic risk model can serve as an independent prognostic factor, guiding the diagnosis and treatment of NSCLC. CEBPA may serve as a potential tumor biomarker and immune target, facilitating further exploration of the biological functions and immunological relevance in NSCLC.
探讨 ARGs 对 NSCLC 预后的影响及其与临床病理参数和免疫微环境的相关性。初步研究 CEBPA 在 NSCLC 中的生物学功能。利用共识聚类分析确定 NSCLC 患者中 ARGs 的分子亚型;利用 LASSO 回归和多变量 Cox 分析选择 7 个预后风险基因并构建预后风险模型;利用森林图分析验证 NSCLC 的独立预后因素;利用 ESTIMATE 和 ssGSEA 分析免疫微环境相关性;通过 qPCR 和 Western 印迹评估 NSCLC 中预后风险基因之间的相关性;使用 CCK-8 和 EdU 检测法测量 NSCLC 中基因敲除和过表达 CEBPA 的 mRNA 和蛋白表达水平;使用 Transwell 实验评估基因敲除和过表达 CEBPA 对细胞增殖的影响;使用 mIHC 分析检测 CEBPA 与 T 细胞和 B 细胞的相关性。共识聚类分析确定了三种分子亚型,表明这些ARGs在NSCLC预后和临床病理参数中的表达存在显著差异。在预后风险模型中,两个风险组之间存在明显的表达差异,P<0.001。预后风险模型的风险评分也是P<0.001。CEBPA 在 NSCLC 细胞系中表现出较高的 mRNA 和蛋白表达水平。siRNA介导的CEBPA基因敲除明显抑制了NSCLC细胞的增殖、迁移和侵袭,而CEBPA的过表达则表现出相反的趋势。mIHC结果显示,CEBPA高表达组的CD3 + CD4+、CD3 + CD8+和CD20 +细胞数明显增加。预后风险模型的风险评分可作为一个独立的预后因素,指导 NSCLC 的诊断和治疗。CEBPA可能是一种潜在的肿瘤生物标记物和免疫靶点,有助于进一步探索其在NSCLC中的生物学功能和免疫学意义。
{"title":"Integrative analysis of aging-related genes reveals CEBPA as a novel therapeutic target in non-small cell lung cancer","authors":"Jiaqi Zhu, Xiaoren Zhu, Conglin Shi, Qixuan Li, Yun Jiang, Xingyou Chen, Pingping Sun, Yi Jin, Tianyi Wang, Jianle Chen","doi":"10.1186/s12935-024-03457-4","DOIUrl":"https://doi.org/10.1186/s12935-024-03457-4","url":null,"abstract":"To explore the impact of ARGs on the prognosis of NSCLC, and its correlation with clinicopathological parameters and immune microenvironment. Preliminary research on the biological functions of CEBPA in NSCLC. Using consensus clustering analysis to identify molecular subtypes of ARGs in NSCLC patients; employing LASSO regression and multivariate Cox analysis to select 7 prognostic risk genes and construct a prognostic risk model; validating independent prognostic factors of NSCLC using forest plot analysis; analyzing immune microenvironment correlations using ESTIMATE and ssGSEA; assessing correlations between prognostic risk genes via qPCR and Western blot in NSCLC; measuring mRNA and protein expression levels of knocked down and overexpressed CEBPA in NSCLC using CCK-8 and EdU assays; evaluating the effects of knocked down and overexpressed CEBPA on cell proliferation using Transwell experiments; examining the correlation of CEBPA with T cells and B cells using mIHC analysis. Consensus clustering analysis identified three molecular subtypes, suggesting significant differential expression of these ARGs in NSCLC prognosis and clinical pathological parameters. There was significant differential expression between the two risk groups in the prognostic risk model, with P < 0.001. The risk score of the prognostic risk model was also P < 0.001. CEBPA exhibited higher mRNA and protein expression levels in NSCLC cell lines. Knockdown of CEBPA significantly reduced mRNA and protein expression levels of CEBPB, YWHAZ, ABL1, and CDK1 in H1650 and A549 cells. siRNA-mediated knockdown of CEBPA markedly inhibited proliferation, migration, and invasion of NSCLC cells, whereas overexpression of CEBPA showed the opposite trend. mIHC results indicated a significant increase in CD3 + CD4+, CD3 + CD8+, and CD20 + cell counts in the high CEBPA expression group. The risk score of the prognostic risk model can serve as an independent prognostic factor, guiding the diagnosis and treatment of NSCLC. CEBPA may serve as a potential tumor biomarker and immune target, facilitating further exploration of the biological functions and immunological relevance in NSCLC.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141785024","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
Cancer Cell International
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
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