首页 > 最新文献

Oncology Research最新文献

英文 中文
Inflammatory myofibroblastic tumor from molecular diagnostics to current treatment. 从分子诊断到当前治疗的炎性肌纤维母细胞瘤。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.050350
Paulina Chmiel, Aleksandra SłOWIKOWSKA, Łukasz Banaszek, Anna Szumera-CIEćKIEWICZ, BARTłOMIEJ Szostakowski, Mateusz J SPAłEK, Tomasz Świtaj, Piotr Rutkowski, Anna M Czarnecka

Inflammatory myofibroblastic tumor (IMT) is a rare neoplasm with intermediate malignancy characterized by a propensity for recurrence but a low metastatic rate. Diagnostic challenges arise from the diverse pathological presentation, variable symptomatology, and lack of different imaging features. However, IMT is identified by the fusion of the anaplastic lymphoma kinase (ALK) gene, which is present in approximately 70% of cases, with various fusion partners, including ran-binding protein 2 (RANBP2), which allows confirmation of the diagnosis. While surgery is the preferred approach for localized tumors, the optimal long-term treatment for advanced or metastatic disease is difficult to define. Targeted therapies are crucial for achieving sustained response to treatment within the context of genetic alteration in IMT. Crizotinib, an ALK tyrosine kinase inhibitor (TKI), was officially approved by the US Food and Drug Administration (FDA) in 2020 to treat IMT with ALK rearrangement. However, most patients face resistance and disease progression, requiring consideration of sequential treatments. Combining radiotherapy with targeted therapy appears to be beneficial in this indication. Early promising results have also been achieved with immunotherapy, indicating potential for combined therapy approaches. However, defined recommendations are still lacking. This review analyzes the available research on IMT, including genetic disorders and their impact on the course of the disease, data on the latest targeted therapy regimens and the possibility of developing immunotherapy in this indication, as well as summarizing general knowledge about prognostic and predictive factors, also in terms of resistance to systemic therapy.

炎性肌纤维母细胞瘤(IMT)是一种罕见的中度恶性肿瘤,其特点是易复发但转移率低。由于病理表现多样、症状多变且缺乏不同的影像学特征,因此诊断难度很大。不过,IMT 可通过无性淋巴瘤激酶(ALK)基因与各种融合伙伴(包括 RAN 结合蛋白 2 (RANBP2))的融合来鉴别,约 70% 的病例存在这种融合,因此可以确诊。虽然手术是治疗局部肿瘤的首选方法,但晚期或转移性疾病的最佳长期治疗方法却难以确定。在 IMT 基因发生改变的情况下,靶向疗法对于实现持续的治疗反应至关重要。克唑替尼是一种ALK酪氨酸激酶抑制剂(TKI),于2020年获得美国食品药品管理局(FDA)的正式批准,用于治疗ALK重排的IMT。然而,大多数患者面临耐药和疾病进展的问题,需要考虑连续治疗。在这一适应症中,放疗与靶向治疗相结合似乎是有益的。早期的免疫疗法也取得了可喜的成果,显示了联合治疗方法的潜力。然而,目前仍缺乏明确的建议。本综述分析了现有的 IMT 研究,包括遗传性疾病及其对病程的影响、最新靶向治疗方案的数据以及在该适应症中开发免疫疗法的可能性,并总结了有关预后和预测因素的一般知识,包括对系统疗法的耐药性。
{"title":"Inflammatory myofibroblastic tumor from molecular diagnostics to current treatment.","authors":"Paulina Chmiel, Aleksandra SłOWIKOWSKA, Łukasz Banaszek, Anna Szumera-CIEćKIEWICZ, BARTłOMIEJ Szostakowski, Mateusz J SPAłEK, Tomasz Świtaj, Piotr Rutkowski, Anna M Czarnecka","doi":"10.32604/or.2024.050350","DOIUrl":"10.32604/or.2024.050350","url":null,"abstract":"<p><p>Inflammatory myofibroblastic tumor (IMT) is a rare neoplasm with intermediate malignancy characterized by a propensity for recurrence but a low metastatic rate. Diagnostic challenges arise from the diverse pathological presentation, variable symptomatology, and lack of different imaging features. However, IMT is identified by the fusion of the anaplastic lymphoma kinase (ALK) gene, which is present in approximately 70% of cases, with various fusion partners, including ran-binding protein 2 (RANBP2), which allows confirmation of the diagnosis. While surgery is the preferred approach for localized tumors, the optimal long-term treatment for advanced or metastatic disease is difficult to define. Targeted therapies are crucial for achieving sustained response to treatment within the context of genetic alteration in IMT. Crizotinib, an ALK tyrosine kinase inhibitor (TKI), was officially approved by the US Food and Drug Administration (FDA) in 2020 to treat IMT with ALK rearrangement. However, most patients face resistance and disease progression, requiring consideration of sequential treatments. Combining radiotherapy with targeted therapy appears to be beneficial in this indication. Early promising results have also been achieved with immunotherapy, indicating potential for combined therapy approaches. However, defined recommendations are still lacking. This review analyzes the available research on IMT, including genetic disorders and their impact on the course of the disease, data on the latest targeted therapy regimens and the possibility of developing immunotherapy in this indication, as well as summarizing general knowledge about prognostic and predictive factors, also in terms of resistance to systemic therapy.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11209743/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469838","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
IKIP downregulates THBS1/FAK signaling to suppress migration and invasion by glioblastoma cells. IKIP 可下调 THBS1/FAK 信号,从而抑制胶质母细胞瘤细胞的迁移和侵袭。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.042456
Zhaoying Zhu, Yanjia Hu, Feng Ye, Haibo Teng, Guoliang You, Yunhui Zeng, Meng Tian, Jianguo Xu, Jin Li, Zhiyong Liu, Hao Liu, Niandong Zheng

Background: Inhibitor of NF-κB kinase-interacting protein (IKIP) is known to promote proliferation of glioblastoma (GBM) cells, but how it affects migration and invasion by those cells is unclear.

Methods: We compared levels of IKIP between glioma tissues and normal brain tissue in clinical samples and public databases. We examined the effects of IKIP overexpression and knockdown on the migration and invasion of GBM using transwell and wound healing assays, and we compared the transcriptomes under these different conditions to identify the molecular mechanisms involved.

Results: Based on data from our clinical samples and from public databases, IKIP was overexpressed in GBM tumors, and its expression level correlated inversely with survival. IKIP overexpression in GBM cells inhibited migration and invasion in transwell and wound healing assays, whereas IKIP knockdown exerted the opposite effects. IKIP overexpression in GBM cells that were injected into mouse brain promoted tumor growth but inhibited tumor invasion of surrounding tissue. The effects of IKIP were associated with downregulation of THBS1 mRNA and concomitant inhibition of THBS1/FAK signaling.

Conclusions: IKIP inhibits THBS1/FAK signaling to suppress migration and invasion of GBM cells.

背景:已知NF-κB激酶相互作用蛋白抑制剂(IKIP)可促进胶质母细胞瘤(GBM)细胞的增殖,但它如何影响这些细胞的迁移和侵袭尚不清楚:我们比较了临床样本和公共数据库中胶质瘤组织和正常脑组织的 IKIP 水平。方法:我们比较了临床样本和公共数据库中神经胶质瘤组织和正常脑组织中 IKIP 的水平,并使用跨孔试验和伤口愈合试验研究了 IKIP 过表达和敲除对神经胶质瘤迁移和侵袭的影响,还比较了不同条件下的转录组,以确定其中的分子机制:结果:根据我们的临床样本和公共数据库的数据,IKIP在GBM肿瘤中过表达,其表达水平与生存率成反比。IKIP在GBM细胞中的过表达抑制了跨孔和伤口愈合实验中的迁移和侵袭,而IKIP的敲除则产生了相反的效果。在注射到小鼠脑部的 GBM 细胞中过表达 IKIP 会促进肿瘤生长,但会抑制肿瘤对周围组织的侵袭。IKIP的作用与THBS1 mRNA的下调以及THBS1/FAK信号传导的抑制有关:结论:IKIP可抑制THBS1/FAK信号转导,从而抑制GBM细胞的迁移和侵袭。
{"title":"IKIP downregulates THBS1/FAK signaling to suppress migration and invasion by glioblastoma cells.","authors":"Zhaoying Zhu, Yanjia Hu, Feng Ye, Haibo Teng, Guoliang You, Yunhui Zeng, Meng Tian, Jianguo Xu, Jin Li, Zhiyong Liu, Hao Liu, Niandong Zheng","doi":"10.32604/or.2024.042456","DOIUrl":"10.32604/or.2024.042456","url":null,"abstract":"<p><strong>Background: </strong>Inhibitor of NF-κB kinase-interacting protein (IKIP) is known to promote proliferation of glioblastoma (GBM) cells, but how it affects migration and invasion by those cells is unclear.</p><p><strong>Methods: </strong>We compared levels of IKIP between glioma tissues and normal brain tissue in clinical samples and public databases. We examined the effects of IKIP overexpression and knockdown on the migration and invasion of GBM using transwell and wound healing assays, and we compared the transcriptomes under these different conditions to identify the molecular mechanisms involved.</p><p><strong>Results: </strong>Based on data from our clinical samples and from public databases, IKIP was overexpressed in GBM tumors, and its expression level correlated inversely with survival. IKIP overexpression in GBM cells inhibited migration and invasion in transwell and wound healing assays, whereas IKIP knockdown exerted the opposite effects. IKIP overexpression in GBM cells that were injected into mouse brain promoted tumor growth but inhibited tumor invasion of surrounding tissue. The effects of IKIP were associated with downregulation of THBS1 mRNA and concomitant inhibition of THBS1/FAK signaling.</p><p><strong>Conclusions: </strong>IKIP inhibits THBS1/FAK signaling to suppress migration and invasion of GBM cells.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11211642/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469837","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
ROR2 promotes invasion and chemoresistance of triple-negative breast cancer cells by activating PI3K/AKT/mTOR signaling. ROR2 通过激活 PI3K/AKT/mTOR 信号,促进三阴性乳腺癌细胞的侵袭和化疗抵抗。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.045433
Xia DA, Han Ge, Junfeng Shi, Chunhua Zhu, Guozhu Wang, Yuan Fang, Jin Xu

Objective: This study aimed to investigate the role of receptor tyrosine kinase-like orphan receptor 2 (ROR2) in triple-negative breast cancer (TNBC).

Methods: ROR2 expression in primary TNBC and metastatic TNBC tissues was analyzed by immunohistochemical staining and PCR. ROR2 expression in TNBC cell lines was detected by PCR and Western blot analysis. The migration, invasion and chemosensitivity of TNBC cells with overexpression or knockdown of ROR2 were examined.

Results: ROR2 expression was high in metastatic TNBC tissues. ROR2 knockdown suppressed the migration, invasion and chemoresistance of TNBC cells. ROR2 overexpression in MDA-MB-435 cells promoted the migration, invasion, and chemoresistance. Moreover, ROR2 knockdown in HC1599 and MDA-MB-435 adriamycin-resistant cells enhanced chemosensitivity to adriamycin. ROR2 could activate PI3K/AKT/mTOR signaling in TNBC cells.

Conclusion: ROR2 is upregulated and promotes metastatic phenotypes of TNBC by activating PI3K/AKT/mTOR signaling.

研究目的本研究旨在探讨受体酪氨酸激酶样孤儿受体2(ROR2)在三阴性乳腺癌(TNBC)中的作用:方法:通过免疫组化染色和 PCR 分析原发性 TNBC 和转移性 TNBC 组织中 ROR2 的表达。通过 PCR 和 Western 印迹分析检测了 ROR2 在 TNBC 细胞系中的表达。检测了过表达或敲除 ROR2 的 TNBC 细胞的迁移、侵袭和化疗敏感性:结果:ROR2在转移性TNBC组织中高表达。结果:ROR2在转移性TNBC组织中高表达,ROR2敲除抑制了TNBC细胞的迁移、侵袭和化疗耐受性。ROR2 在 MDA-MB-435 细胞中的过表达促进了细胞的迁移、侵袭和耐药性。此外,在HC1599和MDA-MB-435阿霉素耐药细胞中敲除ROR2可增强细胞对阿霉素的化疗敏感性。ROR2可激活TNBC细胞的PI3K/AKT/mTOR信号转导:结论:ROR2上调并通过激活PI3K/AKT/mTOR信号促进TNBC的转移表型。
{"title":"ROR2 promotes invasion and chemoresistance of triple-negative breast cancer cells by activating PI3K/AKT/mTOR signaling.","authors":"Xia DA, Han Ge, Junfeng Shi, Chunhua Zhu, Guozhu Wang, Yuan Fang, Jin Xu","doi":"10.32604/or.2024.045433","DOIUrl":"10.32604/or.2024.045433","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to investigate the role of receptor tyrosine kinase-like orphan receptor 2 (ROR2) in triple-negative breast cancer (TNBC).</p><p><strong>Methods: </strong>ROR2 expression in primary TNBC and metastatic TNBC tissues was analyzed by immunohistochemical staining and PCR. ROR2 expression in TNBC cell lines was detected by PCR and Western blot analysis. The migration, invasion and chemosensitivity of TNBC cells with overexpression or knockdown of ROR2 were examined.</p><p><strong>Results: </strong>ROR2 expression was high in metastatic TNBC tissues. ROR2 knockdown suppressed the migration, invasion and chemoresistance of TNBC cells. ROR2 overexpression in MDA-MB-435 cells promoted the migration, invasion, and chemoresistance. Moreover, ROR2 knockdown in HC1599 and MDA-MB-435 adriamycin-resistant cells enhanced chemosensitivity to adriamycin. ROR2 could activate PI3K/AKT/mTOR signaling in TNBC cells.</p><p><strong>Conclusion: </strong>ROR2 is upregulated and promotes metastatic phenotypes of TNBC by activating PI3K/AKT/mTOR signaling.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11209745/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469840","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Trametinib boosts palbociclib's efficacy in breast cancer via autophagy inhibition. 曲美替尼通过抑制自噬增强帕博西尼对乳腺癌的疗效
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.046139
Anguo Wu, Jiao Yan, Ting Su, Chi Feng, Xin Long, Yiru Pan, Rupei Ye, Tian Xia, Hanan Long, Jianming Wu, Xiuli Xiao

Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL's inhibitory effects on breast cancer using both in vitro (MCF7 and MDA-MB-468 cells) and in vivo (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL's anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and in vivo studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.

乳腺癌是全球主要的健康问题,需要不断探索新的治疗策略。帕博西尼(Palbociclib,PAL)是一种著名的细胞周期蛋白依赖性激酶(CDK)抑制剂,在乳腺癌治疗中发挥着至关重要的作用。尽管其疗效已得到认可,但人们对 PAL 与细胞自噬之间的相互作用,尤其是在 RAF/MEK/ERK 信号通路背景下的相互作用,仍然缺乏足够的探索。本研究利用体外(MCF7 和 MDA-MB-468 细胞)和体内(肿瘤裸鼠)模型研究了 PAL 对乳腺癌的抑制作用。我们的研究旨在阐明PAL对自噬过程的影响,并探索其与MEK抑制剂曲美替尼(TRA)联用的潜力,以应对PAL诱导的耐药性挑战。我们的研究结果表明,PAL 能显著降低 MCF7 和 MDA-MB-468 细胞的存活率,缩小小鼠肿瘤的大小,同时对 MCF10A 细胞的细胞毒性极小。然而,PAL 还能诱导保护性自噬,可能通过激活 RAF/MEK/ERK 通路导致耐药性。引入 TRA 能有效中和这种自噬,增强 PAL 的抗肿瘤功效。PAL 和 TRA 的组合能协同降低细胞活力和增殖,体内研究显示肿瘤明显缩小。总之,PAL 和 TRA 的组合是克服 PAL 诱导的耐药性的一种有前途的策略,为乳腺癌治疗开辟了新天地。
{"title":"Trametinib boosts palbociclib's efficacy in breast cancer via autophagy inhibition.","authors":"Anguo Wu, Jiao Yan, Ting Su, Chi Feng, Xin Long, Yiru Pan, Rupei Ye, Tian Xia, Hanan Long, Jianming Wu, Xiuli Xiao","doi":"10.32604/or.2024.046139","DOIUrl":"10.32604/or.2024.046139","url":null,"abstract":"<p><p>Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL's inhibitory effects on breast cancer using both <i>in vitro</i> (MCF7 and MDA-MB-468 cells) and <i>in vivo</i> (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL's anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and <i>in vivo</i> studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11209742/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
LncRNA PCGEM1 facilitates cervical cancer progression via miR-642a-5p/KIF5B axis. LncRNA PCGEM1通过miR-642a-5p/KIF5B轴促进宫颈癌的进展。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.047454
Yuanlin Liu, Yan Liu, Yan Wang, Qiang Wang, Yan Yan, Dandan Zhang, Huiqin Liu

At present, the role of many long non-coding RNAs (lncRNAs) as tumor suppressors in the formation and development of cervical cancer (CC) has been studied. However, lncRNA prostate cancer gene expression marker 1 (PCGEM1), whose high expression not only aggravates ovarian cancer but also can induce tumorigenesis and endometrial cancer progression, has not been studied in CC. The objective of this study was to investigate the expression and the underlying role of PCGEM1 in CC. The relative expression of PCGEM1 in CC cells was detected by real-time PCR. After the suppression of PCGEM1 expression by shRNA, the changes in the proliferation, migration, and invasion capacities were detected via CCK-8 assay, EdU assay, and colony formation assay wound healing assay. Transwell assay and the changes in expressions of epithelial-to-mesenchymal transition (EMT) markers were determined by western blot and immunofluorescence. The interplay among PCGEM1, miR-642a-5p, and kinesin family member 5B (KIF5B) was confirmed by bioinformatics analyses and luciferase reporter assay. Results showed that PCGEM1 expressions were up-regulated within CC cells. Cell viabilities, migration, and invasion were remarkably reduced after the suppression of PCGEM1 expression by shRNA in Hela and SiHa cells. N-cadherin was silenced, but E-cadherin expression was elevated by sh-PCGEM1. Moreover, by sponging miR-642a-5p in CC, PCGEM1 was verified as a competitive endogenous RNA (ceRNA) that modulates KIF5B levels. MiR-642a-5p down-regulation partially rescued sh-PCGEM1's inhibitory effects on cell proliferation, migration, invasion, and EMT process. In conclusion, the PCGEM1/miR-642a-5p/KIF5B signaling axis might be a novel therapeutic target in CC. This study provides a research basis and new direction for targeted therapy of CC.

目前,许多长非编码 RNA(lncRNA)作为肿瘤抑制因子在宫颈癌(CC)的形成和发展中的作用已被研究。然而,lncRNA前列腺癌基因表达标志物1(PCGEM1)的高表达不仅会加重卵巢癌的病情,还能诱导肿瘤发生和子宫内膜癌的进展,但目前尚未对其在CC中的作用进行研究。本研究旨在探讨 PCGEM1 在 CC 中的表达及其潜在作用。研究采用实时 PCR 技术检测 PCGEM1 在 CC 细胞中的相对表达。用 shRNA 抑制 PCGEM1 表达后,通过 CCK-8 试验、EdU 试验和集落形成试验检测细胞增殖、迁移和侵袭能力的变化。通过 Western 印迹和免疫荧光检测了 Transwell 试验和上皮细胞向间质转化(EMT)标志物表达的变化。生物信息学分析和荧光素酶报告实验证实了PCGEM1、miR-642a-5p和驱动蛋白家族成员5B(KIF5B)之间的相互作用。结果表明,PCGEM1 的表达在 CC 细胞中上调。用 shRNA 抑制 Hela 和 SiHa 细胞中 PCGEM1 的表达后,细胞活力、迁移和侵袭能力显著降低。sh-PCGEM1抑制了N-cadherin的表达,但提高了E-cadherin的表达。此外,通过在CC中加入miR-642a-5p,PCGEM1被证实是一种竞争性内源性RNA(ceRNA),可调节KIF5B的水平。下调 MiR-642a-5p 可部分缓解 sh-PCGEM1 对细胞增殖、迁移、侵袭和 EMT 过程的抑制作用。总之,PCGEM1/miR-642a-5p/KIF5B信号轴可能是CC的一个新的治疗靶点。这项研究为CC的靶向治疗提供了研究基础和新方向。
{"title":"LncRNA PCGEM1 facilitates cervical cancer progression via miR-642a-5p/KIF5B axis.","authors":"Yuanlin Liu, Yan Liu, Yan Wang, Qiang Wang, Yan Yan, Dandan Zhang, Huiqin Liu","doi":"10.32604/or.2024.047454","DOIUrl":"10.32604/or.2024.047454","url":null,"abstract":"<p><p>At present, the role of many long non-coding RNAs (lncRNAs) as tumor suppressors in the formation and development of cervical cancer (CC) has been studied. However, lncRNA prostate cancer gene expression marker 1 (PCGEM1), whose high expression not only aggravates ovarian cancer but also can induce tumorigenesis and endometrial cancer progression, has not been studied in CC. The objective of this study was to investigate the expression and the underlying role of PCGEM1 in CC. The relative expression of PCGEM1 in CC cells was detected by real-time PCR. After the suppression of PCGEM1 expression by shRNA, the changes in the proliferation, migration, and invasion capacities were detected via CCK-8 assay, EdU assay, and colony formation assay wound healing assay. Transwell assay and the changes in expressions of epithelial-to-mesenchymal transition (EMT) markers were determined by western blot and immunofluorescence. The interplay among PCGEM1, miR-642a-5p, and kinesin family member 5B (KIF5B) was confirmed by bioinformatics analyses and luciferase reporter assay. Results showed that PCGEM1 expressions were up-regulated within CC cells. Cell viabilities, migration, and invasion were remarkably reduced after the suppression of PCGEM1 expression by shRNA in Hela and SiHa cells. N-cadherin was silenced, but E-cadherin expression was elevated by sh-PCGEM1. Moreover, by sponging miR-642a-5p in CC, PCGEM1 was verified as a competitive endogenous RNA (ceRNA) that modulates KIF5B levels. MiR-642a-5p down-regulation partially rescued sh-PCGEM1's inhibitory effects on cell proliferation, migration, invasion, and EMT process. In conclusion, the PCGEM1/miR-642a-5p/KIF5B signaling axis might be a novel therapeutic target in CC. This study provides a research basis and new direction for targeted therapy of CC.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11209744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469839","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Silencing of the long non-coding RNA LINC00265 triggers autophagy and apoptosis in lung cancer by reducing protein stability of SIN3A oncogene. 沉默长非编码 RNA LINC00265 可通过降低 SIN3A 癌基因蛋白的稳定性,引发肺癌自噬和细胞凋亡。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2023.030771
Xiaobi Huang, Chunyuan Chen, Yongyang Chen, Honglian Zhou, Yonghua Chen, Zhong Huang, Yuliu Xie, Baiyang Liu, Yudong Guo, Zhixiong Yang, Guanghua Chen, Wenmei Su

Background: Long non-coding RNAs are important regulators in cancer biology and function either as tumor suppressors or as oncogenes. Their dysregulation has been closely associated with tumorigenesis. LINC00265 is upregulated in lung adenocarcinoma and is a prognostic biomarker of this cancer. However, the mechanism underlying its function in cancer progression remains poorly understood.

Methods: Here, the regulatory role of LINC00265 in lung adenocarcinoma was examined using lung cancer cell lines, clinical samples, and xenografts.

Results: We found that high levels of LINC00265 expression were associated with shorter overall survival rate of patients, whereas knockdown of LINC00265 inhibited proliferation of cancer cell lines and tumor growth in xenografts. Western blot and flow cytometry analyses indicated that silencing of LINC00265 induced autophagy and apoptosis. Moreover, we showed that LINC00265 interacted with and stabilized the transcriptional co-repressor Switch-independent 3a (SIN3A), which is a scaffold protein functioning either as a tumor repressor or as an oncogene in a context-dependent manner. Silencing of SIN3A also reduced proliferation of lung cancer cells, which was correlated with the induction of autophagy. These observations raise the possibility that LINC00265 functions to promote the oncogenic activity of SIN3A in lung adenocarcinoma.

Conclusions: Our findings thus identify SIN3A as a LINC00265-associated protein and should help to understand the mechanism underlying LINC00265-mediated oncogenesis.

背景:长非编码 RNA 是癌症生物学中的重要调控因子,可作为肿瘤抑制因子或致癌基因发挥作用。它们的失调与肿瘤发生密切相关。LINC00265 在肺腺癌中上调,是这种癌症的预后生物标志物。方法:本文利用肺癌细胞系、临床样本和异种移植物研究了 LINC00265 在肺腺癌中的调控作用:结果:我们发现LINC00265的高水平表达与患者较短的总生存率有关,而LINC00265的敲除抑制了癌细胞株的增殖和异种移植物的肿瘤生长。Western印迹和流式细胞术分析表明,沉默LINC00265可诱导自噬和细胞凋亡。此外,我们还发现,LINC00265 与转录共抑制因子开关独立 3a(SIN3A)相互作用并使其稳定,SIN3A 是一种支架蛋白,其功能要么是肿瘤抑制因子,要么是致癌基因,其作用方式取决于具体情况。沉默 SIN3A 还能减少肺癌细胞的增殖,这与诱导自噬有关。这些观察结果表明,LINC00265有可能在肺腺癌中促进SIN3A的致癌活性:因此,我们的研究结果确定了 SIN3A 是一种 LINC00265 相关蛋白,这将有助于了解 LINC00265 介导的致癌机制。
{"title":"Silencing of the long non-coding RNA <i>LINC00265</i> triggers autophagy and apoptosis in lung cancer by reducing protein stability of SIN3A oncogene.","authors":"Xiaobi Huang, Chunyuan Chen, Yongyang Chen, Honglian Zhou, Yonghua Chen, Zhong Huang, Yuliu Xie, Baiyang Liu, Yudong Guo, Zhixiong Yang, Guanghua Chen, Wenmei Su","doi":"10.32604/or.2023.030771","DOIUrl":"10.32604/or.2023.030771","url":null,"abstract":"<p><strong>Background: </strong>Long non-coding RNAs are important regulators in cancer biology and function either as tumor suppressors or as oncogenes. Their dysregulation has been closely associated with tumorigenesis. <i>LINC00265</i> is upregulated in lung adenocarcinoma and is a prognostic biomarker of this cancer. However, the mechanism underlying its function in cancer progression remains poorly understood.</p><p><strong>Methods: </strong>Here, the regulatory role of <i>LINC00265</i> in lung adenocarcinoma was examined using lung cancer cell lines, clinical samples, and xenografts.</p><p><strong>Results: </strong>We found that high levels of <i>LINC00265</i> expression were associated with shorter overall survival rate of patients, whereas knockdown of <i>LINC00265</i> inhibited proliferation of cancer cell lines and tumor growth in xenografts. Western blot and flow cytometry analyses indicated that silencing of <i>LINC00265</i> induced autophagy and apoptosis. Moreover, we showed that <i>LINC00265</i> interacted with and stabilized the transcriptional co-repressor Switch-independent 3a (SIN3A), which is a scaffold protein functioning either as a tumor repressor or as an oncogene in a context-dependent manner. Silencing of SIN3A also reduced proliferation of lung cancer cells, which was correlated with the induction of autophagy. These observations raise the possibility that <i>LINC00265</i> functions to promote the oncogenic activity of SIN3A in lung adenocarcinoma.</p><p><strong>Conclusions: </strong>Our findings thus identify SIN3A as a <i>LINC00265</i>-associated protein and should help to understand the mechanism underlying <i>LINC00265</i>-mediated oncogenesis.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11211643/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
The effect of celastrol in combination with 5-fluorouracil on proliferation and apoptosis of gastric cancer cell lines. 青霉烯醇与 5-氟尿嘧啶联用对胃癌细胞株增殖和凋亡的影响
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-20 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.047187
Mohammad-Taghi Moradi, Dhiya Altememy, Majid Asadi-Samani, Pegah Khosravian, Marziyeh Soltani, Leila Hashemi, Azadeh Samiei-Sefat

Background: Despite the availability of chemotherapy drugs such as 5-fluorouracil (5-FU), the treatment of some cancers such as gastric cancer remains challenging due to drug resistance and side effects. This study aimed to investigate the effect of celastrol in combination with the chemotherapy drug 5-FU on proliferation and induction of apoptosis in human gastric cancer cell lines (AGS and EPG85-257).

Materials and methods: In this in vitro study, AGS and EPG85-257 cells were treated with different concentrations of celastrol, 5-FU, and their combination. Cell proliferation was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The synergistic effect of 5-FU and celastrol was studied using Compusyn software. The DNA content at different phases of the cell cycle and apoptosis rate was measured using flow cytometry.

Results: Co-treatment with low concentrations (10% inhibitory concentration (IC10)) of celastrol and 5-FU significantly reduced IC50 (p < 0.05) so that 48 h after treatment, IC50 was calculated at 3.77 and 6.9 μM for celastrol, 20.7 and 11.6 μM for 5-FU, and 5.03 and 4.57 μM for their combination for AGS and EPG85-257 cells, respectively. The mean percentage of apoptosis for AGS cells treated with celastrol, 5-FU, and their combination was obtained 23.9, 41.2, and 61.9, and for EPG85-257 cells 5.65, 46.9, and 55.7, respectively. In addition, the 5-FU and celastrol-5-FU combination induced cell cycle arrest in the synthesis phase.

Conclusions: Although celastrol could decrease the concentration of 5-fluorouracil that sufficed to suppress gastric cancer cells, additional studies are required to arrive at conclusive evidence on the anticancer effects of celastrol.

背景:尽管有5-氟尿嘧啶(5-FU)等化疗药物,但由于耐药性和副作用,一些癌症(如胃癌)的治疗仍面临挑战。本研究旨在探讨青霉烯醇与化疗药物 5-FU 联用对人胃癌细胞株(AGS 和 EPG85-257)增殖和诱导凋亡的影响:在这项体外研究中,AGS 和 EPG85-257 细胞分别接受了不同浓度的西司他醇、5-FU 和它们的联合治疗。细胞增殖采用 3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四氮唑(MTT)检测法进行评估。使用 Compusyn 软件研究了 5-FU 和青霉烯醇的协同效应。使用流式细胞术测量了细胞周期不同阶段的 DNA 含量和细胞凋亡率:结果:低浓度(10%抑制浓度(IC10))的青霉烯醇和 5-FU 联合处理可显著降低 IC50(p < 0.05),因此处理 48 小时后,计算出 AGS 和 EPG85-257 细胞的 IC50 分别为 3.77 和 6.9 μM,5-FU 分别为 20.7 和 11.6 μM,它们的组合分别为 5.03 和 4.57 μM。经芹甾醇、5-FU 和它们的组合处理的 AGS 细胞的平均凋亡百分比分别为 23.9、41.2 和 61.9,EPG85-257 细胞的平均凋亡百分比分别为 5.65、46.9 和 55.7。此外,5-FU 和青霉烯醇-5-FU 组合可诱导细胞周期停滞在合成期:结论:虽然青霉烯醇能降低足以抑制胃癌细胞的 5-氟尿嘧啶浓度,但还需要更多的研究才能得出青霉烯醇抗癌作用的确凿证据。
{"title":"The effect of celastrol in combination with 5-fluorouracil on proliferation and apoptosis of gastric cancer cell lines.","authors":"Mohammad-Taghi Moradi, Dhiya Altememy, Majid Asadi-Samani, Pegah Khosravian, Marziyeh Soltani, Leila Hashemi, Azadeh Samiei-Sefat","doi":"10.32604/or.2024.047187","DOIUrl":"10.32604/or.2024.047187","url":null,"abstract":"<p><strong>Background: </strong>Despite the availability of chemotherapy drugs such as 5-fluorouracil (5-FU), the treatment of some cancers such as gastric cancer remains challenging due to drug resistance and side effects. This study aimed to investigate the effect of celastrol in combination with the chemotherapy drug 5-FU on proliferation and induction of apoptosis in human gastric cancer cell lines (AGS and EPG85-257).</p><p><strong>Materials and methods: </strong>In this <i>in vitro</i> study, AGS and EPG85-257 cells were treated with different concentrations of celastrol, 5-FU, and their combination. Cell proliferation was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The synergistic effect of 5-FU and celastrol was studied using Compusyn software. The DNA content at different phases of the cell cycle and apoptosis rate was measured using flow cytometry.</p><p><strong>Results: </strong>Co-treatment with low concentrations (10% inhibitory concentration (IC10)) of celastrol and 5-FU significantly reduced IC50 (<i>p</i> < 0.05) so that 48 h after treatment, IC50 was calculated at 3.77 and 6.9 μM for celastrol, 20.7 and 11.6 μM for 5-FU, and 5.03 and 4.57 μM for their combination for AGS and EPG85-257 cells, respectively. The mean percentage of apoptosis for AGS cells treated with celastrol, 5-FU, and their combination was obtained 23.9, 41.2, and 61.9, and for EPG85-257 cells 5.65, 46.9, and 55.7, respectively. In addition, the 5-FU and celastrol-5-FU combination induced cell cycle arrest in the synthesis phase.</p><p><strong>Conclusions: </strong>Although celastrol could decrease the concentration of 5-fluorouracil that sufficed to suppress gastric cancer cells, additional studies are required to arrive at conclusive evidence on the anticancer effects of celastrol.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11209740/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469842","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
GNAS mutations suppress cell invasion by activating MEG3 in growth hormone-secreting pituitary adenoma. 在分泌生长激素的垂体腺瘤中,GNAS突变通过激活MEG3抑制细胞侵袭。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-05-23 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.046007
Chao Tang, Chunyu Zhong, Junhao Zhu, Feng Yuan, Jin Yang, Yong Xu, Chiyuan Ma

Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in GNAS (α subunit of stimulatory G protein). Mutations in GNAS are associated with clinical features of smaller and less invasive tumors. However, the role of GNAS mutations in the invasiveness of GHPAs is unclear. GNAS mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (MEG3) was evaluated with RT-qPCR. MEG3 was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of MEG3 on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated GNAS compared with that in mice with wild-type GNAS. Consistently, the invasiveness of mutant GNAS-expressing GH3 cells decreased. MEG3 is uniquely expressed at high levels in GHPAs harboring mutated GNAS. Accordingly, MEG3 upregulation inhibited tumor cell invasion, and conversely, MEG3 downregulation increased tumor cell invasion. Mechanistically, GNAS mutations inhibit EMT in GHPAs. MEG3 in mutated GNAS cells prevented cell invasion through the inactivation of the Wnt/β-catenin signaling pathway, which was further validated in vivo. Our data suggest that GNAS mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the MEG3/Wnt/β-catenin signaling pathway.

大约30%-40%的分泌生长激素的垂体腺瘤(GHPAs)携带GNAS(刺激性G蛋白α亚基)的体细胞激活突变。GNAS 突变与肿瘤较小、侵袭性较低的临床特征有关。然而,GNAS突变在GHPA侵袭性中的作用尚不清楚。采用标准聚合酶链反应(PCR)测序程序检测了GHPA中的GNAS突变。用 RT-qPCR 评估了与突变相关的母体表达基因 3(MEG3)的表达。使用慢病毒表达系统对 GH3 细胞中的 MEG3 进行操作。使用 Transwell 试验测定细胞侵袭能力,并通过免疫荧光和 Western 印迹对上皮-间质转化(EMT)相关蛋白进行定量。最后,使用肿瘤细胞异种移植小鼠模型来验证 MEG3 对肿瘤生长和侵袭性的影响。与野生型GNAS小鼠相比,突变型GNAS小鼠的GHPA侵袭性明显降低。同样,表达突变 GNAS 的 GH3 细胞的侵袭性也降低了。在携带突变型 GNAS 的 GHPA 中,MEG3 独特地高水平表达。因此,MEG3 的上调抑制了肿瘤细胞的侵袭,反之,MEG3 的下调增加了肿瘤细胞的侵袭。从机理上讲,GNAS突变抑制了GHPAs的EMT。突变的GNAS细胞中的MEG3通过使Wnt/β-catenin信号通路失活来阻止细胞侵袭,这在体内得到了进一步验证。我们的数据表明,GNAS突变可能通过激活MEG3/Wnt/β-catenin信号通路调节EMT,从而抑制GHPAs的细胞侵袭。
{"title":"<i>GNAS</i> mutations suppress cell invasion by activating MEG3 in growth hormone-secreting pituitary adenoma.","authors":"Chao Tang, Chunyu Zhong, Junhao Zhu, Feng Yuan, Jin Yang, Yong Xu, Chiyuan Ma","doi":"10.32604/or.2024.046007","DOIUrl":"10.32604/or.2024.046007","url":null,"abstract":"<p><p>Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in <i>GNAS</i> (α subunit of stimulatory G protein). Mutations in <i>GNAS</i> are associated with clinical features of smaller and less invasive tumors. However, the role of <i>GNAS</i> mutations in the invasiveness of GHPAs is unclear. <i>GNAS</i> mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (<i>MEG3</i>) was evaluated with RT-qPCR. <i>MEG3</i> was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of <i>MEG3</i> on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated <i>GNAS</i> compared with that in mice with wild-type <i>GNAS</i>. Consistently, the invasiveness of mutant <i>GNAS</i>-expressing GH3 cells decreased. <i>MEG3</i> is uniquely expressed at high levels in GHPAs harboring mutated <i>GNAS</i>. Accordingly, <i>MEG3</i> upregulation inhibited tumor cell invasion, and conversely, <i>MEG3</i> downregulation increased tumor cell invasion. Mechanistically, <i>GNAS</i> mutations inhibit EMT in GHPAs. <i>MEG3</i> in mutated <i>GNAS</i> cells prevented cell invasion through the inactivation of the Wnt/β-catenin signaling pathway, which was further validated <i>in vivo</i>. Our data suggest that <i>GNAS</i> mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the <i>MEG3/Wnt/β-catenin</i> signaling pathway.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11136687/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141198988","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
New insights into ATR inhibition in muscle invasive bladder cancer: The role of apolipoprotein B mRNA editing catalytic subunit 3B. 肌肉浸润性膀胱癌 ATR 抑制的新见解:脂蛋白 B mRNA 编辑催化亚基 3B 的作用。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-05-23 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.048919
Hyunho Kim, Uiju Cho, Sook Hee Hong, Hyung Soon Park, In-Ho Kim, Ho Jung An, Byoung Yong Shim, Jin Hyoung Kang

Background: Apolipoprotein B mRNA editing catalytic polypeptide (APOBEC), an endogenous mutator, induces DNA damage and activates the ataxia telangiectasia and Rad3-related (ATR)-checkpoint kinase 1 (Chk1) pathway. Although cisplatin-based therapy is the mainstay for muscle-invasive bladder cancer (MIBC), it has a poor survival rate. Therefore, this study aimed to evaluate the efficacy of an ATR inhibitor combined with cisplatin in the treatment of APOBEC catalytic subunit 3B (APOBEC3B) expressing MIBC.

Methods: Immunohistochemical staining was performed to analyze an association between APOBEC3B and ATR in patients with MIBC. The APOBEC3B expression in MIBC cell lines was assessed using real-time polymerase chain reaction and western blot analysis. Western blot analysis was performed to confirm differences in phosphorylated Chk1 (pChk1) expression according to the APOBEC3B expression. Cell viability and apoptosis analyses were performed to examine the anti-tumor activity of ATR inhibitors combined with cisplatin.

Conclusion: There was a significant association between APOBEC3B and ATR expression in the tumor tissues obtained from patients with MIBC. Cells with higher APOBEC3B expression showed higher pChk1 expression than cells expressing low APOBEC3B levels. Combination treatment of ATR inhibitor and cisplatin inhibited cell growth in MIBC cells with a higher APOBEC3B expression. Compared to cisplatin single treatment, combination treatment induced more apoptotic cell death in the cells with higher APOBEC3B expression. Conclusion: Our study shows that APOBEC3B's higher expression status can enhance the sensitivity of MIBC to cisplatin upon ATR inhibition. This result provides new insight into appropriate patient selection for the effective application of ATR inhibitors in MIBC.

背景:载脂蛋白B mRNA编辑催化多肽(APOBEC)是一种内源性突变体,可诱导DNA损伤并激活共济失调毛细血管扩张和Rad3相关(ATR)-检查点激酶1(Chk1)通路。尽管顺铂疗法是治疗肌浸润性膀胱癌(MIBC)的主要方法,但其生存率较低。因此,本研究旨在评估ATR抑制剂联合顺铂治疗表达APOBEC催化亚基3B(APOBEC3B)的MIBC的疗效:方法:对MIBC患者进行免疫组化染色,分析APOBEC3B与ATR之间的关联。使用实时聚合酶链反应和 Western 印迹分析评估了 APOBEC3B 在 MIBC 细胞系中的表达。根据 APOBEC3B 的表达情况,进行了 Western 印迹分析以确认磷酸化 Chk1(pChk1)表达的差异。进行了细胞活力和细胞凋亡分析,以检验ATR抑制剂联合顺铂的抗肿瘤活性:结论:在MIBC患者的肿瘤组织中,APOBEC3B和ATR的表达有明显的关联。APOBEC3B表达较高的细胞比APOBEC3B表达较低的细胞显示出更高的pChk1表达。ATR 抑制剂和顺铂联合治疗可抑制 APOBEC3B 表达较高的 MIBC 细胞的生长。与顺铂单一治疗相比,联合治疗在 APOBEC3B 表达较高的细胞中诱导了更多的细胞凋亡。结论我们的研究表明,APOBEC3B的高表达状态可提高ATR抑制后MIBC对顺铂的敏感性。这一结果为ATR抑制剂在MIBC中的有效应用提供了新的见解。
{"title":"New insights into ATR inhibition in muscle invasive bladder cancer: The role of apolipoprotein B mRNA editing catalytic subunit 3B.","authors":"Hyunho Kim, Uiju Cho, Sook Hee Hong, Hyung Soon Park, In-Ho Kim, Ho Jung An, Byoung Yong Shim, Jin Hyoung Kang","doi":"10.32604/or.2024.048919","DOIUrl":"10.32604/or.2024.048919","url":null,"abstract":"<p><strong>Background: </strong>Apolipoprotein B mRNA editing catalytic polypeptide (APOBEC), an endogenous mutator, induces DNA damage and activates the ataxia telangiectasia and Rad3-related (ATR)-checkpoint kinase 1 (Chk1) pathway. Although cisplatin-based therapy is the mainstay for muscle-invasive bladder cancer (MIBC), it has a poor survival rate. Therefore, this study aimed to evaluate the efficacy of an ATR inhibitor combined with cisplatin in the treatment of APOBEC catalytic subunit 3B (APOBEC3B) expressing MIBC.</p><p><strong>Methods: </strong>Immunohistochemical staining was performed to analyze an association between APOBEC3B and ATR in patients with MIBC. The APOBEC3B expression in MIBC cell lines was assessed using real-time polymerase chain reaction and western blot analysis. Western blot analysis was performed to confirm differences in phosphorylated Chk1 (pChk1) expression according to the APOBEC3B expression. Cell viability and apoptosis analyses were performed to examine the anti-tumor activity of ATR inhibitors combined with cisplatin.</p><p><strong>Conclusion: </strong>There was a significant association between APOBEC3B and ATR expression in the tumor tissues obtained from patients with MIBC. Cells with higher APOBEC3B expression showed higher pChk1 expression than cells expressing low APOBEC3B levels. Combination treatment of ATR inhibitor and cisplatin inhibited cell growth in MIBC cells with a higher APOBEC3B expression. Compared to cisplatin single treatment, combination treatment induced more apoptotic cell death in the cells with higher APOBEC3B expression. <b>Conclusion:</b> Our study shows that APOBEC3B's higher expression status can enhance the sensitivity of MIBC to cisplatin upon ATR inhibition. This result provides new insight into appropriate patient selection for the effective application of ATR inhibitors in MIBC.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":2.0,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11136685/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141199475","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
MicroRNAs in thyroid cancer with focus on medullary thyroid carcinoma: potential therapeutic targets and diagnostic/prognostic markers and web based tools. 甲状腺癌中的微RNAs,重点是甲状腺髓样癌:潜在的治疗靶点和诊断/预后标志物以及基于网络的工具。
IF 3.1 4区 医学 Q1 Medicine Pub Date : 2024-05-23 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.049235
Elham Shakiba, Seti Boroomand, Sima Kheradmand Kia, Mehdi Hedayati

This review aimed to describe the inculpation of microRNAs (miRNAs) in thyroid cancer (TC) and its subtypes, mainly medullary thyroid carcinoma (MTC), and to outline web-based tools and databases for bioinformatics analysis of miRNAs in TC. Additionally, the capacity of miRNAs to serve as therapeutic targets and biomarkers in TC management will be discussed. This review is based on a literature search of relevant articles on the role of miRNAs in TC and its subtypes, mainly MTC. Additionally, web-based tools and databases for bioinformatics analysis of miRNAs in TC were identified and described. MiRNAs can perform as oncomiRs or antioncoges, relying on the target mRNAs they regulate. MiRNA replacement therapy using miRNA mimics or antimiRs that aim to suppress the function of certain miRNAs can be applied to correct miRNAs aberrantly expressed in diseases, particularly in cancer. MiRNAs are involved in the modulation of fundamental pathways related to cancer, resembling cell cycle checkpoints and DNA repair pathways. MiRNAs are also rather stable and can reliably be detected in different types of biological materials, rendering them favorable diagnosis and prognosis biomarkers as well. MiRNAs have emerged as promising tools for evaluating medical outcomes in TC and as possible therapeutic targets. The contribution of miRNAs in thyroid cancer, particularly MTC, is an active area of research, and the utility of web applications and databases for the biological data analysis of miRNAs in TC is becoming increasingly important.

本综述旨在描述微RNA(miRNA)在甲状腺癌(TC)及其亚型(主要是甲状腺髓样癌(MTC))中的应用,并概述基于网络的TC中miRNA生物信息学分析工具和数据库。此外,还将讨论 miRNA 作为治疗靶点和生物标志物在甲状腺癌治疗中的作用。本综述基于对有关 miRNA 在 TC 及其亚型(主要是 MTC)中作用的相关文章的文献检索。此外,还发现并介绍了用于对 TC 中 miRNA 进行生物信息学分析的网络工具和数据库。miRNA可作为oncomiRs或antioncoges发挥作用,这取决于它们调控的靶mRNA。使用旨在抑制某些 miRNA 功能的 miRNA 模拟物或 antimiRs 进行 MiRNA 替代治疗,可用于纠正疾病(尤其是癌症)中异常表达的 miRNA。miRNA 参与了与癌症有关的基本通路的调节,如细胞周期检查点和 DNA 修复通路。MiRNA 也相当稳定,可以在不同类型的生物材料中可靠地检测到,因此也是有利的诊断和预后生物标志物。MiRNA 已成为评估 TC 医疗结果的有前途的工具,并可能成为治疗靶点。miRNA在甲状腺癌(尤其是MTC)中的作用是一个活跃的研究领域,网络应用程序和数据库在甲状腺癌miRNA生物数据分析中的应用正变得越来越重要。
{"title":"MicroRNAs in thyroid cancer with focus on medullary thyroid carcinoma: potential therapeutic targets and diagnostic/prognostic markers and web based tools.","authors":"Elham Shakiba, Seti Boroomand, Sima Kheradmand Kia, Mehdi Hedayati","doi":"10.32604/or.2024.049235","DOIUrl":"10.32604/or.2024.049235","url":null,"abstract":"<p><p>This review aimed to describe the inculpation of microRNAs (miRNAs) in thyroid cancer (TC) and its subtypes, mainly medullary thyroid carcinoma (MTC), and to outline web-based tools and databases for bioinformatics analysis of miRNAs in TC. Additionally, the capacity of miRNAs to serve as therapeutic targets and biomarkers in TC management will be discussed. This review is based on a literature search of relevant articles on the role of miRNAs in TC and its subtypes, mainly MTC. Additionally, web-based tools and databases for bioinformatics analysis of miRNAs in TC were identified and described. MiRNAs can perform as oncomiRs or antioncoges, relying on the target mRNAs they regulate. MiRNA replacement therapy using miRNA mimics or antimiRs that aim to suppress the function of certain miRNAs can be applied to correct miRNAs aberrantly expressed in diseases, particularly in cancer. MiRNAs are involved in the modulation of fundamental pathways related to cancer, resembling cell cycle checkpoints and DNA repair pathways. MiRNAs are also rather stable and can reliably be detected in different types of biological materials, rendering them favorable diagnosis and prognosis biomarkers as well. MiRNAs have emerged as promising tools for evaluating medical outcomes in TC and as possible therapeutic targets. The contribution of miRNAs in thyroid cancer, particularly MTC, is an active area of research, and the utility of web applications and databases for the biological data analysis of miRNAs in TC is becoming increasingly important.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-05-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11136686/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141199400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
期刊
Oncology Research
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