首页 > 最新文献

Cancer Biology & Therapy最新文献

英文 中文
WYC-209 inhibited GC malignant progression by down-regulating WNT4 through RARα. WYC-209 通过 RARα 下调 WNT4,从而抑制 GC 的恶性发展。
IF 3.6 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-01-04 DOI: 10.1080/15384047.2023.2299288
Zhenyuan Qian, Wenfa Lin, Xufan Cai, Jianzhang Wu, Kun Ke, Zaiyuan Ye, Fang Wu

Gastric cancer (GC) has been a major health burden all over the world but there are fewer promising chemotherapeutic drugs due to its multidrug resistance. It has been reported that WYC-209 suppresses the growth and metastasis of tumor-repopulating cells but the effect on GC was not explored. MTT, colony formation, and transwell assays were performed to examine the effects of WYC-209 on the proliferation, colony growth, and mobility of GC cells. Western blotting and qRT-PCR were used to detect the expression of proteins and mRNA. RNA-seq and enrichment analyses were conducted for the differentially expressed genes and enriched biological processes and pathways. The rescue experiments were carried out for further validation. Besides, we constructed xenograft model to confirm the effect of WYC-209 in vivo. The dual-luciferase reporter and Chromatin immunoprecipitation were implemented to confirm the underlying mechanism. WYC-209 exerted excellent anti-cancer effects both in vitro and in vivo. Based on RNA-seq and enrichment analyses, we found that Wnt family member 4 (WNT4) was significantly down-regulated. More importantly, WNT4 overexpression breached the inhibitory effect of WYC-209 on GC progression. Mechanically, WYC-209 significantly promoted the binding between retinoic acid receptor α (RARα) and WNT4 promoter. WYC-209 exerts anti-tumor effects in GC by down-regulating the expression of WNT4 via RARα.

胃癌(GC)一直是全球主要的健康负担,但由于其多药耐药性,有前景的化疗药物较少。有报道称,WYC-209 可抑制肿瘤繁殖细胞的生长和转移,但对 GC 的影响尚未探究。为了研究 WYC-209 对 GC 细胞的增殖、集落生长和移动性的影响,我们进行了 MTT、集落形成和透孔试验。采用 Western 印迹和 qRT-PCR 检测蛋白质和 mRNA 的表达。对差异表达基因和富集的生物过程和通路进行了 RNA-seq 和富集分析。为了进一步验证,我们还进行了拯救实验。此外,我们还构建了异种移植模型来证实 WYC-209 在体内的作用。我们还利用双荧光素酶报告和染色质免疫沉淀来证实其潜在机制。WYC-209在体外和体内都发挥了很好的抗癌作用。基于 RNA-seq 和富集分析,我们发现 Wnt 家族成员 4(WNT4)被显著下调。更重要的是,WNT4的过表达破坏了WYC-209对GC进展的抑制作用。在机制上,WYC-209 能明显促进视黄酸受体α(RARα)与 WNT4 启动子的结合。WYC-209 通过 RARα 下调 WNT4 的表达,从而在 GC 中发挥抗肿瘤作用。
{"title":"WYC-209 inhibited GC malignant progression by down-regulating WNT4 through RARα.","authors":"Zhenyuan Qian, Wenfa Lin, Xufan Cai, Jianzhang Wu, Kun Ke, Zaiyuan Ye, Fang Wu","doi":"10.1080/15384047.2023.2299288","DOIUrl":"10.1080/15384047.2023.2299288","url":null,"abstract":"<p><p>Gastric cancer (GC) has been a major health burden all over the world but there are fewer promising chemotherapeutic drugs due to its multidrug resistance. It has been reported that WYC-209 suppresses the growth and metastasis of tumor-repopulating cells but the effect on GC was not explored. MTT, colony formation, and transwell assays were performed to examine the effects of WYC-209 on the proliferation, colony growth, and mobility of GC cells. Western blotting and qRT-PCR were used to detect the expression of proteins and mRNA. RNA-seq and enrichment analyses were conducted for the differentially expressed genes and enriched biological processes and pathways. The rescue experiments were carried out for further validation. Besides, we constructed xenograft model to confirm the effect of WYC-209 in vivo. The dual-luciferase reporter and Chromatin immunoprecipitation were implemented to confirm the underlying mechanism. WYC-209 exerted excellent anti-cancer effects both in vitro and in vivo. Based on RNA-seq and enrichment analyses, we found that Wnt family member 4 (WNT4) was significantly down-regulated. More importantly, WNT4 overexpression breached the inhibitory effect of WYC-209 on GC progression. Mechanically, WYC-209 significantly promoted the binding between retinoic acid receptor α (RARα) and WNT4 promoter. WYC-209 exerts anti-tumor effects in GC by down-regulating the expression of WNT4 via RARα.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2299288"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10773637/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139097377","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
Epigenetic silencing ZSCAN23 promotes pancreatic cancer growth by activating Wnt signaling. 表观遗传沉默 ZSCAN23 通过激活 Wnt 信号促进胰腺癌生长
IF 3.6 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-01-16 DOI: 10.1080/15384047.2024.2302924
Qian Du, Meiying Zhang, Aiai Gao, Tao He, Mingzhou Guo

Pancreatic ductal adenocarcinoma (PDAC) is the most malignant tumor. Zinc finger and SCAN domain-containing protein 23 (ZSCAN23) is a new member of the SCAN domain family. The expression regulation and biological function remain to be elucidated. In this study, we explored the epigenetic regulation and the function of ZSCAN23 in PDAC. ZSCAN23 was methylated in 60.21% (171/284) of PDAC and its expression was regulated by promoter region methylation. The expression of ZSCAN23 inhibited cell proliferation, colony formation, migration, invasion, and induced apoptosis and G1/S phase arrest. ZSCAN23 suppressed Panc10.05 cell xenograft growth in mice. Mechanistically, ZSCAN23 inhibited Wnt signaling by interacting with myosin heavy chain 9 (MYH9) in pancreatic cancer cells. ZSCAN23 is frequently methylated in PDAC and may serve as a detective marker. ZSCAN23 suppresses PDAC cell growth both in vitro and in vivo.

胰腺导管腺癌(PDAC)是恶性程度最高的肿瘤。锌指和含 SCAN 结构域蛋白 23(ZSCAN23)是 SCAN 结构域家族的新成员。其表达调控和生物学功能仍有待阐明。本研究探讨了ZSCAN23在PDAC中的表观遗传调控及其功能。在60.21%(171/284)的PDAC中,ZSCAN23被甲基化,其表达受启动子区甲基化调控。ZSCAN23的表达可抑制细胞增殖、集落形成、迁移和侵袭,并诱导细胞凋亡和G1/S期停滞。ZSCAN23抑制了Panc10.05细胞在小鼠体内的异种移植生长。从机理上讲,ZSCAN23通过与胰腺癌细胞中的肌球蛋白重链9(MYH9)相互作用来抑制Wnt信号转导。ZSCAN23在PDAC中经常发生甲基化,可作为一种检测标记物。ZSCAN23 在体外和体内都能抑制 PDAC 细胞的生长。
{"title":"Epigenetic silencing <i>ZSCAN23</i> promotes pancreatic cancer growth by activating Wnt signaling.","authors":"Qian Du, Meiying Zhang, Aiai Gao, Tao He, Mingzhou Guo","doi":"10.1080/15384047.2024.2302924","DOIUrl":"10.1080/15384047.2024.2302924","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is the most malignant tumor. Zinc finger and SCAN domain-containing protein 23 (<i>ZSCAN23</i>) is a new member of the SCAN domain family. The expression regulation and biological function remain to be elucidated. In this study, we explored the epigenetic regulation and the function of <i>ZSCAN23</i> in PDAC. <i>ZSCAN23</i> was methylated in 60.21% (171/284) of PDAC and its expression was regulated by promoter region methylation. The expression of <i>ZSCAN23</i> inhibited cell proliferation, colony formation, migration, invasion, and induced apoptosis and G1/S phase arrest. <i>ZSCAN23</i> suppressed Panc10.05 cell xenograft growth in mice. Mechanistically, <i>ZSCAN23</i> inhibited Wnt signaling by interacting with myosin heavy chain 9 (MYH9) in pancreatic cancer cells. <i>ZSCAN23</i> is frequently methylated in PDAC and may serve as a detective marker. <i>ZSCAN23</i> suppresses PDAC cell growth both <i>in vitro</i> and <i>in vivo</i>.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2302924"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10793710/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139472315","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
M6A modification regulates tumor suppressor DIRAS1 expression in cervical cancer cells. M6A 修饰调节宫颈癌细胞中肿瘤抑制因子 DIRAS1 的表达。
IF 3.6 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-02-19 DOI: 10.1080/15384047.2024.2306674
Yu-Yan Wang, Lian-Hua Ye, An-Qi Zhao, Wei-Ran Gao, Ning Dai, Yu Yin, Xin Zhang

DIRAS family GTPase 1 (DIRAS1) has been reported as a potential tumor suppressor in other human cancer. However, its expression pattern and role in cervical cancer remain unknown. Knockdown of DIRAS1 significantly promoted the proliferation, growth, migration, and invasion of C33A and SiHa cells cultured in vitro. Overexpression of DIRAS1 significantly inhibited the viability and motility of C33A and SiHa cells. Compared with normal cervical tissues, DIRAS1 mRNA levels were significantly lower in cervical cancer tissues. DIRAS1 protein expression was also significantly reduced in cervical cancer tissues compared with para-cancerous tissues. In addition, DIRAS1 expression level in tumor tissues was significantly negatively correlated with the pathological grades of cervical cancer patients. DNA methylation inhibitor (5-Azacytidine) and histone deacetylation inhibitor (SAHA) resulted in a significant increase in DIRAS1 mRNA levels in C33A and SiHa cells, but did not affect DIRAS1 protein levels. FTO inhibitor (FB23-2) significantly down-regulated intracellular DIRAS1 mRNA levels, but significantly up-regulated DIRAS1 protein levels. Moreover, the down-regulation of METTL3 and METTL14 expression significantly inhibited DIRAS1 protein expression, whereas the down-regulation of FTO and ALKBH5 expression significantly increased DIRAS1 protein expression. In conclusion, DIRAS1 exerts a significant anti-oncogenic function and its expression is significantly downregulated in cervical cancer cells. The m6A modification may be a key mechanism to regulate DIRAS1 mRNA stability and protein translation efficiency in cervical cancer.

据报道,DIRAS 家族 GTPase 1(DIRAS1)在其他人类癌症中是一种潜在的肿瘤抑制因子。然而,它在宫颈癌中的表达模式和作用仍然未知。敲除 DIRAS1 能显著促进体外培养的 C33A 和 SiHa 细胞的增殖、生长、迁移和侵袭。过表达 DIRAS1 能明显抑制 C33A 和 SiHa 细胞的活力和运动。与正常宫颈组织相比,宫颈癌组织中的 DIRAS1 mRNA 水平明显较低。与癌旁组织相比,宫颈癌组织中 DIRAS1 蛋白表达也明显降低。此外,肿瘤组织中 DIRAS1 的表达水平与宫颈癌患者的病理分级呈明显负相关。DNA 甲基化抑制剂(5-氮杂胞嘧啶)和组蛋白去乙酰化抑制剂(SAHA)会导致 C33A 和 SiHa 细胞中 DIRAS1 mRNA 水平的显著增加,但不会影响 DIRAS1 蛋白水平。FTO 抑制剂(FB23-2)可显著下调细胞内 DIRAS1 mRNA 水平,但可显著上调 DIRAS1 蛋白水平。此外,下调 METTL3 和 METTL14 的表达可明显抑制 DIRAS1 蛋白的表达,而下调 FTO 和 ALKBH5 的表达可明显增加 DIRAS1 蛋白的表达。总之,DIRAS1 在宫颈癌细胞中具有明显的抗癌功能,且其表达明显下调。m6A 修饰可能是调控宫颈癌中 DIRAS1 mRNA 稳定性和蛋白翻译效率的关键机制。
{"title":"M6A modification regulates tumor suppressor DIRAS1 expression in cervical cancer cells.","authors":"Yu-Yan Wang, Lian-Hua Ye, An-Qi Zhao, Wei-Ran Gao, Ning Dai, Yu Yin, Xin Zhang","doi":"10.1080/15384047.2024.2306674","DOIUrl":"10.1080/15384047.2024.2306674","url":null,"abstract":"<p><p>DIRAS family GTPase 1 (DIRAS1) has been reported as a potential tumor suppressor in other human cancer. However, its expression pattern and role in cervical cancer remain unknown. Knockdown of DIRAS1 significantly promoted the proliferation, growth, migration, and invasion of C33A and SiHa cells cultured <i>in vitro</i>. Overexpression of DIRAS1 significantly inhibited the viability and motility of C33A and SiHa cells. Compared with normal cervical tissues, DIRAS1 mRNA levels were significantly lower in cervical cancer tissues. DIRAS1 protein expression was also significantly reduced in cervical cancer tissues compared with para-cancerous tissues. In addition, DIRAS1 expression level in tumor tissues was significantly negatively correlated with the pathological grades of cervical cancer patients. DNA methylation inhibitor (5-Azacytidine) and histone deacetylation inhibitor (SAHA) resulted in a significant increase in DIRAS1 mRNA levels in C33A and SiHa cells, but did not affect DIRAS1 protein levels. FTO inhibitor (FB23-2) significantly down-regulated intracellular DIRAS1 mRNA levels, but significantly up-regulated DIRAS1 protein levels. Moreover, the down-regulation of METTL3 and METTL14 expression significantly inhibited DIRAS1 protein expression, whereas the down-regulation of FTO and ALKBH5 expression significantly increased DIRAS1 protein expression. In conclusion, DIRAS1 exerts a significant anti-oncogenic function and its expression is significantly downregulated in cervical cancer cells. The m6A modification may be a key mechanism to regulate DIRAS1 mRNA stability and protein translation efficiency in cervical cancer.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2306674"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10878024/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139899398","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
Association between the IL1B-511 C>T polymorphism and the risk of hematologic malignancies: data from a meta-analysis. IL1B-511 C>T 多态性与血液系统恶性肿瘤风险之间的关系:一项荟萃分析的数据。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-07-22 DOI: 10.1080/15384047.2024.2382503
Fabíola Silva Alves-Hanna, Felipe Rodolfo Pereira Silva, Daniele Sá Pereira, Alessandro Luiz Araújo Bentes Leal, Fábio Magalhães-Gama, Allyson Guimarães Costa

The relationship between the IL1B-511C>T (rs16944) polymorphism and the risk of developing hematologic malignancies remains controversial. Thus, we performed a meta-analysis to evaluate the association between IL1B-511C>T polymorphism and the risk of developing hematologic malignancies. A comprehensive search was conducted to identify all eligible studies on IL1B-511C>T polymorphism and hematologic malignancies. Twelve case-control studies, with 2,896 cases and 3,716 controls, were selected for the analysis. The overall data failed to indicate a significant association between IL1B-511C>T polymorphism and the risk of hematologic malignancies (OR:1.06, 95% Confidence Interval [CI]: 0.93-1.22). Moreover, non-significant associations were observed in a stratified analysis according to neoplasm type (multiple myeloma, Hodgkin's lymphoma, and non-Hodgkin's lymphoma), ethnicity (European and Asian), and Hardy-Weinberg equilibrium. In summary, our results suggest that there is no association between the IL1B-511C>T polymorphism and the risk of hematologic malignancies. As such, further large-scale studies are needed to confirm our findings.

IL1B-511C>T(rs16944)多态性与罹患血液系统恶性肿瘤风险之间的关系仍存在争议。因此,我们进行了一项荟萃分析,以评估IL1B-511C>T多态性与罹患血液系统恶性肿瘤风险之间的关系。我们进行了一次全面检索,以确定所有符合条件的关于IL1B-511C>T多态性与血液恶性肿瘤的研究。分析选取了 12 项病例对照研究,包括 2,896 例病例和 3,716 例对照。总体数据未能表明IL1B-511C>T多态性与血液系统恶性肿瘤风险之间存在显著关联(OR:1.06,95% 置信区间[CI]:0.93-1.22)。此外,在根据肿瘤类型(多发性骨髓瘤、霍奇金淋巴瘤和非霍奇金淋巴瘤)、种族(欧洲人和亚洲人)和哈代-温伯格平衡进行的分层分析中,也观察到了不显著的相关性。总之,我们的研究结果表明,IL1B-511C>T 多态性与罹患血液系统恶性肿瘤的风险之间没有关联。因此,还需要进一步的大规模研究来证实我们的发现。
{"title":"Association between the <i>IL1B-511 C>T</i> polymorphism and the risk of hematologic malignancies: data from a meta-analysis.","authors":"Fabíola Silva Alves-Hanna, Felipe Rodolfo Pereira Silva, Daniele Sá Pereira, Alessandro Luiz Araújo Bentes Leal, Fábio Magalhães-Gama, Allyson Guimarães Costa","doi":"10.1080/15384047.2024.2382503","DOIUrl":"10.1080/15384047.2024.2382503","url":null,"abstract":"<p><p>The relationship between the <i>IL1B-511C>T</i> (rs16944) polymorphism and the risk of developing hematologic malignancies remains controversial. Thus, we performed a meta-analysis to evaluate the association between <i>IL1B-511C>T</i> polymorphism and the risk of developing hematologic malignancies. A comprehensive search was conducted to identify all eligible studies on <i>IL1B-511C>T</i> polymorphism and hematologic malignancies. Twelve case-control studies, with 2,896 cases and 3,716 controls, were selected for the analysis. The overall data failed to indicate a significant association between <i>IL1B-511C>T</i> polymorphism and the risk of hematologic malignancies (OR:1.06, 95% Confidence Interval [CI]: 0.93-1.22). Moreover, non-significant associations were observed in a stratified analysis according to neoplasm type (multiple myeloma, Hodgkin's lymphoma, and non-Hodgkin's lymphoma), ethnicity (European and Asian), and Hardy-Weinberg equilibrium. In summary, our results suggest that there is no association between the <i>IL1B-511C>T</i> polymorphism and the risk of hematologic malignancies. As such, further large-scale studies are needed to confirm our findings.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2382503"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11268255/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141747530","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
Mechanism investigation of Forsythoside A against esophageal squamous cell carcinoma in vitro and in vivo. 连翘苷 A 对食管鳞状细胞癌的体内外作用机制研究
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-07-24 DOI: 10.1080/15384047.2024.2380023
Yingying Yang, Junru Shen, Peiyuan Deng, Ping Chen

Context: Forsythoside A (FSA) was extracted from Forsythia suspensa, a traditional Chinese medicine, which has been demonstrated to exert anti-inflammatory, antibacterial, and other pharmacological effects. However, the anticancer effect of FSA in esophageal squamous cell carcinoma (ESCC) has not been documented.

Objective: The present study aimed to elucidate the mechanism of FSA against ESCC.

Materials and methods: Network pharmacology and molecular docking were employed to predict the mechanism. FSA was utilized to treat ESCC cell lines KYSE450 and KYSE30, followed by CCK-8 assay, cell cloning formation assay, flow cytometry, Western blot, RNA-seq analysis, and subsequent in vivo experiments.

Results: Network pharmacology and molecular docking predicted that the therapeutic effect of FSA in ESCC is mediated through proteins such as BCL2 and BAX, influencing KEGG pathways associated with apoptosis. In vitro experiments showed that FSA inhibited cell proliferation and plate clone formation, promoted cell apoptosis and impacted the cell cycle distribution of G2/M phase by regulating BCL2, BAX, and p21. Further RNA-seq in KYSE450 cells showed that FSA regulated the expression of 223 genes, specifically affecting the biological process of epidermal development. In vivo experiments showed that gastric administration of FSA resulted in notable reductions in both tumor volume and weight by regulating BCL2, BAX, and p21. 16S rRNA sequencing showed that FSA led to significant changes of beta diversity. Abundance of 11 specific bacterial taxa were considerably changed following administration of FSA.

Conclusions: This study presents a novel candidate drug against ESCC and establishes a foundation for future clinical application.

背景:连翘苷 A(FSA)是从中药连翘中提取的,已被证实具有抗炎、抗菌和其他药理作用。然而,FSA 对食管鳞状细胞癌(ESCC)的抗癌作用尚未见文献记载:本研究旨在阐明 FSA 对 ESCC 的作用机制:采用网络药理学和分子对接法预测其机制。利用FSA处理ESCC细胞株KYSE450和KYSE30,然后进行CCK-8检测、细胞克隆形成检测、流式细胞术、Western blot、RNA-seq分析和随后的体内实验:网络药理学和分子对接预测,FSA对ESCC的治疗作用是通过BCL2和BAX等蛋白介导的,影响了与细胞凋亡相关的KEGG通路。体外实验表明,FSA通过调控BCL2、BAX和p21抑制细胞增殖和平板克隆形成,促进细胞凋亡,并影响G2/M期的细胞周期分布。进一步的 KYSE450 细胞 RNA 序列分析表明,FSA 可调控 223 个基因的表达,特别是影响表皮发育的生物学过程。体内实验表明,通过调节 BCL2、BAX 和 p21,胃部给药 FSA 可显著减少肿瘤体积和重量。16S rRNA测序显示,FSA导致了β多样性的显著变化。服用 FSA 后,11 个特定细菌类群的丰度发生了显著变化:本研究提出了一种抗 ESCC 的新型候选药物,为未来的临床应用奠定了基础。
{"title":"Mechanism investigation of Forsythoside A against esophageal squamous cell carcinoma <i>in vitro</i> and <i>in vivo</i>.","authors":"Yingying Yang, Junru Shen, Peiyuan Deng, Ping Chen","doi":"10.1080/15384047.2024.2380023","DOIUrl":"10.1080/15384047.2024.2380023","url":null,"abstract":"<p><strong>Context: </strong>Forsythoside A (FSA) was extracted from Forsythia suspensa, a traditional Chinese medicine, which has been demonstrated to exert anti-inflammatory, antibacterial, and other pharmacological effects. However, the anticancer effect of FSA in esophageal squamous cell carcinoma (ESCC) has not been documented.</p><p><strong>Objective: </strong>The present study aimed to elucidate the mechanism of FSA against ESCC.</p><p><strong>Materials and methods: </strong>Network pharmacology and molecular docking were employed to predict the mechanism. FSA was utilized to treat ESCC cell lines KYSE450 and KYSE30, followed by CCK-8 assay, cell cloning formation assay, flow cytometry, Western blot, RNA-seq analysis, and subsequent in vivo experiments.</p><p><strong>Results: </strong>Network pharmacology and molecular docking predicted that the therapeutic effect of FSA in ESCC is mediated through proteins such as BCL2 and BAX, influencing KEGG pathways associated with apoptosis. In vitro experiments showed that FSA inhibited cell proliferation and plate clone formation, promoted cell apoptosis and impacted the cell cycle distribution of G2/M phase by regulating BCL2, BAX, and p21. Further RNA-seq in KYSE450 cells showed that FSA regulated the expression of 223 genes, specifically affecting the biological process of epidermal development. In vivo experiments showed that gastric administration of FSA resulted in notable reductions in both tumor volume and weight by regulating BCL2, BAX, and p21. 16S rRNA sequencing showed that FSA led to significant changes of beta diversity. Abundance of 11 specific bacterial taxa were considerably changed following administration of FSA.</p><p><strong>Conclusions: </strong>This study presents a novel candidate drug against ESCC and establishes a foundation for future clinical application.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2380023"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11271126/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141751184","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
Exploring the molecular mechanisms and therapeutic potential of SMAD4 in colorectal cancer. 探索 SMAD4 在结直肠癌中的分子机制和治疗潜力。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-08-20 DOI: 10.1080/15384047.2024.2392341
Hui Shan, Guangyu Tian, Yeqing Zhang, Zhiyuan Qiu

Colorectal Cancer (CRC) is the third most common cancer worldwide, and the occurrence and development of CRC are influenced by the molecular biology characteristics of CRC, especially alterations in key signaling pathways. The transforming growth factor-β (TGF-β) plays a crucial role in cellular growth, differentiation, migration, and apoptosis, with SMAD4 protein serving as a key transcription factor in the TGF-β signaling pathway, thus playing a significant role in the onset and progression of CRC. CRC is one of the malignancies with a high mortality rate worldwide. Despite significant research progress in recent years, especially regarding the role of SMAD4, its dual role in the early and late stages of tumor progression has promoted further discussion on its complexity as a therapeutic target, highlighting the urgent need for a deeper analysis of its role in CRC. This review aims to explore the function of SMAD4 protein in CRC and its potential as a therapeutic target.

结直肠癌(CRC)是全球第三大常见癌症,CRC的发生和发展受其分子生物学特征的影响,尤其是关键信号通路的改变。转化生长因子-β(TGF-β)在细胞生长、分化、迁移和凋亡中起着至关重要的作用,其中 SMAD4 蛋白是 TGF-β 信号通路中的一个关键转录因子,因此在 CRC 的发病和进展中起着重要作用。CRC 是全球死亡率较高的恶性肿瘤之一。尽管近年来研究取得了重大进展,尤其是关于 SMAD4 的作用,但其在肿瘤进展早期和晚期的双重作用促使人们进一步讨论其作为治疗靶点的复杂性,突出了深入分析其在 CRC 中作用的迫切需要。本综述旨在探讨 SMAD4 蛋白在 CRC 中的功能及其作为治疗靶点的潜力。
{"title":"Exploring the molecular mechanisms and therapeutic potential of SMAD4 in colorectal cancer.","authors":"Hui Shan, Guangyu Tian, Yeqing Zhang, Zhiyuan Qiu","doi":"10.1080/15384047.2024.2392341","DOIUrl":"10.1080/15384047.2024.2392341","url":null,"abstract":"<p><p>Colorectal Cancer (CRC) is the third most common cancer worldwide, and the occurrence and development of CRC are influenced by the molecular biology characteristics of CRC, especially alterations in key signaling pathways. The transforming growth factor-β (TGF-β) plays a crucial role in cellular growth, differentiation, migration, and apoptosis, with SMAD4 protein serving as a key transcription factor in the TGF-β signaling pathway, thus playing a significant role in the onset and progression of CRC. CRC is one of the malignancies with a high mortality rate worldwide. Despite significant research progress in recent years, especially regarding the role of SMAD4, its dual role in the early and late stages of tumor progression has promoted further discussion on its complexity as a therapeutic target, highlighting the urgent need for a deeper analysis of its role in CRC. This review aims to explore the function of SMAD4 protein in CRC and its potential as a therapeutic target.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2392341"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11340766/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008296","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
Role and mechanism of COL3A1 in regulating the growth, metastasis, and drug sensitivity in cisplatin-resistant non-small cell lung cancer cells. COL3A1在调节顺铂耐药非小细胞肺癌细胞的生长、转移和药物敏感性中的作用和机制
IF 3.6 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-03-26 DOI: 10.1080/15384047.2024.2328382
Jiankun Ren, Songwei Zhao, Junyu Lai

Non-small cell lung cancer (NSCLC) is among the most difficult malignancies to treat. Type III collagen (COL3A1) can affect the progression and chemoresistance development of NSCLC. We herein explored the mechanism that drives COL3A1 dysregulation in NSCLC. Potential RNA-binding proteins (RBPs) and transcription factors (TFs) that could bind to COL3A1 were searched by bioinformatics. mRNA expression was detected by quantitative PCR. Protein expression was evaluated using immunoblotting and immunohistochemistry. The effects of the variables were assessed by gauging cell growth, invasiveness, migratory capacity, apoptosis, and cisplatin (DDP) sensitivity. The direct YY1/COL3A1 relationship was confirmed by ChIP and luciferase reporter experiments. Xenograft experiments were done to examine COL3A1's function in DDP efficacy. COL3A1 showed enhanced expression in DDP-resistant NSCLC. In H460/DDP and A549/DDP cells, downregulation of COL3A1 exerted inhibitory functions in cell growth, invasiveness, and migration, as well as promoting effects on cell DDP sensitivity and apoptosis. Mechanistically, ELAV-like RNA binding protein 1 (ELAVL1) enhanced the mRNA stability and expression of COL3A1, and Yin Yang 1 (YY1) promoted the transcription and expression of COL3A1. Furthermore, upregulation of COL3A1 reversed ELAVL1 inhibition- or YY1 deficiency-mediated functions in DDP-resistant NSCLC cells. Additionally, COL3A1 downregulation enhanced the anti-tumor efficacy of DDP in vivo. Our investigation demonstrates that COL3A1 upregulation, induced by both RBP ELAVL1 and TF YY1, exerts important functions in phenotypes of NSCLC cells with DDP resistance, offering an innovative opportunity in the treatment of drug-resistant NSCLC.

非小细胞肺癌(NSCLC)是最难治疗的恶性肿瘤之一。III型胶原蛋白(COL3A1)可影响非小细胞肺癌的进展和化疗耐药性的形成。我们在此探讨了NSCLC中COL3A1失调的驱动机制。通过生物信息学搜索了可能与 COL3A1 结合的潜在 RNA 结合蛋白(RBPs)和转录因子(TFs)。使用免疫印迹法和免疫组织化学法评估蛋白质表达。通过测量细胞生长、侵袭性、迁移能力、细胞凋亡和顺铂(DDP)敏感性来评估变量的影响。通过 ChIP 和荧光素酶报告实验证实了 YY1/COL3A1 的直接关系。异种移植实验检验了 COL3A1 在 DDP 疗效中的功能。COL3A1在对DDP耐药的NSCLC中表达增强。在H460/DDP和A549/DDP细胞中,下调COL3A1可抑制细胞生长、侵袭性和迁移,并促进细胞对DDP的敏感性和凋亡。从机制上看,ELAV样RNA结合蛋白1(ELAVL1)增强了COL3A1的mRNA稳定性和表达,阴阳1(YY1)促进了COL3A1的转录和表达。此外,COL3A1的上调逆转了ELAVL1抑制或YY1缺乏介导的DDP耐药NSCLC细胞的功能。此外,COL3A1的下调增强了DDP在体内的抗肿瘤功效。我们的研究表明,RBP ELAVL1和TF YY1诱导的COL3A1上调在DDP耐药的NSCLC细胞表型中发挥了重要作用,为耐药NSCLC的治疗提供了创新机会。
{"title":"Role and mechanism of COL3A1 in regulating the growth, metastasis, and drug sensitivity in cisplatin-resistant non-small cell lung cancer cells.","authors":"Jiankun Ren, Songwei Zhao, Junyu Lai","doi":"10.1080/15384047.2024.2328382","DOIUrl":"10.1080/15384047.2024.2328382","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) is among the most difficult malignancies to treat. Type III collagen (COL3A1) can affect the progression and chemoresistance development of NSCLC. We herein explored the mechanism that drives COL3A1 dysregulation in NSCLC. Potential RNA-binding proteins (RBPs) and transcription factors (TFs) that could bind to COL3A1 were searched by bioinformatics. mRNA expression was detected by quantitative PCR. Protein expression was evaluated using immunoblotting and immunohistochemistry. The effects of the variables were assessed by gauging cell growth, invasiveness, migratory capacity, apoptosis, and cisplatin (DDP) sensitivity. The direct YY1/COL3A1 relationship was confirmed by ChIP and luciferase reporter experiments. Xenograft experiments were done to examine COL3A1's function in DDP efficacy. COL3A1 showed enhanced expression in DDP-resistant NSCLC. In H460/DDP and A549/DDP cells, downregulation of COL3A1 exerted inhibitory functions in cell growth, invasiveness, and migration, as well as promoting effects on cell DDP sensitivity and apoptosis. Mechanistically, ELAV-like RNA binding protein 1 (ELAVL1) enhanced the mRNA stability and expression of COL3A1, and Yin Yang 1 (YY1) promoted the transcription and expression of COL3A1. Furthermore, upregulation of COL3A1 reversed ELAVL1 inhibition- or YY1 deficiency-mediated functions in DDP-resistant NSCLC cells. Additionally, COL3A1 downregulation enhanced the anti-tumor efficacy of DDP in vivo. Our investigation demonstrates that COL3A1 upregulation, induced by both RBP ELAVL1 and TF YY1, exerts important functions in phenotypes of NSCLC cells with DDP resistance, offering an innovative opportunity in the treatment of drug-resistant NSCLC.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2328382"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10968311/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287029","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
MDFI promotes the proliferation and tolerance to chemotherapy of colorectal cancer cells by binding ITGB4/LAMB3 to activate the AKT signaling pathway. MDFI 通过结合 ITGB4/LAMB3 激活 AKT 信号通路,促进结直肠癌细胞的增殖和对化疗的耐受性。
IF 3.6 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-02-20 DOI: 10.1080/15384047.2024.2314324
Ding Ma, Shuwen Liu, Kua Liu, Lingkai Kong, Lingjun Xiao, Qilei Xin, Chunping Jiang, Junhua Wu

Colorectal cancer (CRC) is one of the most lethal cancers. Single-cell RNA sequencing (scRNA-seq) and protein-protein interactions (PPIs) have enabled the systematic study of CRC. In our research, the activation of the AKT pathway in CRC was analyzed by KEGG using single-cell sequencing data from the GSE144735 dataset. The correlation and PPIs of MDFI and ITGB4/LAMB3 were examined. The results were verified in the TCGA and CCLE and further tested by coimmunoprecipitation experiments. The effect of MDFI on the AKT pathway via ITGB4/LAMB3 was validated by knockdown and lentiviral overexpression experiments. The effect of MDFI on oxaliplatin/fluorouracil sensitivity was probed by colony formation assay and CCK8 assay. We discovered that MDFI was positively associated with ITGB4/LAMB3. In addition, MDFI was negatively associated with oxaliplatin/fluorouracil sensitivity. MDFI upregulated the AKT pathway by directly interacting with LAMB3 and ITGB4 in CRC cells, and enhanced the proliferation of CRC cells via the AKT pathway. Finally, MDFI reduced the sensitivity of CRC cells to oxaliplatin and fluorouracil. In conclusion, MDFI promotes the proliferation and tolerance to chemotherapy of colorectal cancer cells, partially through the activation of the AKT signaling pathway by the binding to ITGB4/LAMB3. Our findings provide a possible molecular target for CRC therapy.

结肠直肠癌(CRC)是致死率最高的癌症之一。单细胞 RNA 测序(scRNA-seq)和蛋白-蛋白相互作用(PPIs)使得对 CRC 的系统研究成为可能。在我们的研究中,利用 GSE144735 数据集中的单细胞测序数据,通过 KEGG 分析了 CRC 中 AKT 通路的激活情况。研究还考察了 MDFI 和 ITGB4/LAMB3 的相关性和 PPIs。这些结果在TCGA和CCLE中得到了验证,并通过共免疫沉淀实验进行了进一步检验。通过敲除和慢病毒过表达实验验证了MDFI通过ITGB4/LAMB3对AKT通路的影响。通过菌落形成实验和CCK8实验检测了MDFI对奥沙利铂/氟尿嘧啶敏感性的影响。我们发现,MDFI与ITGB4/LAMB3呈正相关,与奥沙利铂/氟尿嘧啶敏感性呈负相关。MDFI通过与CRC细胞中的LAMB3和ITGB4直接相互作用,上调AKT通路,并通过AKT通路增强CRC细胞的增殖。最后,MDFI降低了CRC细胞对奥沙利铂和氟尿嘧啶的敏感性。总之,MDFI能促进结直肠癌细胞的增殖和对化疗的耐受性,部分原因是通过与ITGB4/LAMB3结合激活了AKT信号通路。我们的发现为 CRC 治疗提供了一个可能的分子靶点。
{"title":"MDFI promotes the proliferation and tolerance to chemotherapy of colorectal cancer cells by binding ITGB4/LAMB3 to activate the AKT signaling pathway.","authors":"Ding Ma, Shuwen Liu, Kua Liu, Lingkai Kong, Lingjun Xiao, Qilei Xin, Chunping Jiang, Junhua Wu","doi":"10.1080/15384047.2024.2314324","DOIUrl":"10.1080/15384047.2024.2314324","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is one of the most lethal cancers. Single-cell RNA sequencing (scRNA-seq) and protein-protein interactions (PPIs) have enabled the systematic study of CRC. In our research, the activation of the AKT pathway in CRC was analyzed by KEGG using single-cell sequencing data from the GSE144735 dataset. The correlation and PPIs of MDFI and ITGB4/LAMB3 were examined. The results were verified in the TCGA and CCLE and further tested by coimmunoprecipitation experiments. The effect of MDFI on the AKT pathway via ITGB4/LAMB3 was validated by knockdown and lentiviral overexpression experiments. The effect of MDFI on oxaliplatin/fluorouracil sensitivity was probed by colony formation assay and CCK8 assay. We discovered that MDFI was positively associated with ITGB4/LAMB3. In addition, MDFI was negatively associated with oxaliplatin/fluorouracil sensitivity. MDFI upregulated the AKT pathway by directly interacting with LAMB3 and ITGB4 in CRC cells, and enhanced the proliferation of CRC cells via the AKT pathway. Finally, MDFI reduced the sensitivity of CRC cells to oxaliplatin and fluorouracil. In conclusion, MDFI promotes the proliferation and tolerance to chemotherapy of colorectal cancer cells, partially through the activation of the AKT signaling pathway by the binding to ITGB4/LAMB3. Our findings provide a possible molecular target for CRC therapy.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2314324"},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10880501/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139905144","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
PDLIM1, a novel miR-3940-5p target, regulates the malignant progression of diffuse large B-cell lymphoma. 新型 miR-3940-5p 靶点 PDLIM1 可调控弥漫大 B 细胞淋巴瘤的恶性进展。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-11-20 DOI: 10.1080/15384047.2024.2429175
Jinfeng Zhu, Huifang Xiao, Chuntuan Li, Xiaofeng Li, Jinyang Zheng, Xihu Yao, Shaoxiong Wang, Xiongpeng Zhu

Background: PDZ And LIM domain protein 1 (PDLIM1), a protein-coding gene, has been widely reported to exhibit differential expression patterns across various human cancers, including hematological malignancies. This study aimed to investigate PDLIM1 expression pattern and its functional role in diffuse large B-cell lymphoma (DLBCL) both in vitro and in vivo.

Methods: PDLIM1 expression patterns were reanalyzed using data from the Gene Expression Omnibus, and the results were subsequently validated in patient tissue samples and a panel of four DLBCL cell lines. MicroRNA-3940-5p (miR-3940-5p) was identified as an upstream regulator of PDLIM1. The interaction between PDLIM1 and miR-3940-5p and its effects on DLBCL cellular activities and cancer development were further explored using a DLBCL mouse model.

Results: Elevated PDLIM1 expression was observed in DLBCL cells and tissues. Reduced cell proliferation and increased DLBCL cell apoptosis were observed following the knockdown of this gene. Furthermore, short hairpin RNA (shRNA)-mediated PDLIM1 knockdown diminished tumorigenesis of DLBCL cells in nude mice. miR-3940-5p was identified as an upstream regulator of PDLIM1. PDLIM1 expression and function were negatively modulated by the upregulation of miR-3940-5p, consequently affecting the malignant phenotype of DLBCL cells.

Conclusion: These findings suggest that the miR-3940-5p/PDLIM1 axis may play a crucial role in DLBCL pathogenesis and could potentially be exploited for therapeutic interventions.

背景:PDZ 和 LIM 结构域蛋白 1(PDLIM1)是一种蛋白编码基因,已被广泛报道在包括血液恶性肿瘤在内的各种人类癌症中表现出不同的表达模式。本研究旨在研究 PDLIM1 在弥漫大 B 细胞淋巴瘤(DLBCL)中的体外和体内表达模式及其功能作用:方法:利用基因表达总库(Gene Expression Omnibus)中的数据重新分析了PDLIM1的表达模式,随后在患者组织样本和四种DLBCL细胞系中验证了结果。微RNA-3940-5p(miR-3940-5p)被确定为PDLIM1的上游调节因子。利用DLBCL小鼠模型进一步探讨了PDLIM1和miR-3940-5p之间的相互作用及其对DLBCL细胞活性和癌症发展的影响:结果:在DLBCL细胞和组织中观察到PDLIM1表达升高。结果:在 DLBCL 细胞和组织中观察到 PDLIM1 表达升高,敲除该基因后细胞增殖减少,DLBCL 细胞凋亡增加。此外,短发夹核糖核酸(shRNA)介导的 PDLIM1 基因敲除减少了裸鼠 DLBCL 细胞的肿瘤发生。PDLIM1的表达和功能受miR-3940-5p上调的负调控,从而影响DLBCL细胞的恶性表型:这些研究结果表明,miR-3940-5p/PDLIM1 轴可能在 DLBCL 发病机制中起着关键作用,有可能被用于治疗干预。
{"title":"PDLIM1, a novel miR-3940-5p target, regulates the malignant progression of diffuse large B-cell lymphoma.","authors":"Jinfeng Zhu, Huifang Xiao, Chuntuan Li, Xiaofeng Li, Jinyang Zheng, Xihu Yao, Shaoxiong Wang, Xiongpeng Zhu","doi":"10.1080/15384047.2024.2429175","DOIUrl":"10.1080/15384047.2024.2429175","url":null,"abstract":"<p><strong>Background: </strong>PDZ And LIM domain protein 1 (PDLIM1), a protein-coding gene, has been widely reported to exhibit differential expression patterns across various human cancers, including hematological malignancies. This study aimed to investigate PDLIM1 expression pattern and its functional role in diffuse large B-cell lymphoma (DLBCL) both <i>in vitro</i> and <i>in vivo</i>.</p><p><strong>Methods: </strong>PDLIM1 expression patterns were reanalyzed using data from the Gene Expression Omnibus, and the results were subsequently validated in patient tissue samples and a panel of four DLBCL cell lines. MicroRNA-3940-5p (miR-3940-5p) was identified as an upstream regulator of PDLIM1. The interaction between PDLIM1 and miR-3940-5p and its effects on DLBCL cellular activities and cancer development were further explored using a DLBCL mouse model.</p><p><strong>Results: </strong>Elevated PDLIM1 expression was observed in DLBCL cells and tissues. Reduced cell proliferation and increased DLBCL cell apoptosis were observed following the knockdown of this gene. Furthermore, short hairpin RNA (shRNA)-mediated PDLIM1 knockdown diminished tumorigenesis of DLBCL cells in nude mice. miR-3940-5p was identified as an upstream regulator of PDLIM1. PDLIM1 expression and function were negatively modulated by the upregulation of miR-3940-5p, consequently affecting the malignant phenotype of DLBCL cells.</p><p><strong>Conclusion: </strong>These findings suggest that the miR-3940-5p/PDLIM1 axis may play a crucial role in DLBCL pathogenesis and could potentially be exploited for therapeutic interventions.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2429175"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11581179/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142675164","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
Establishment of a visualized mouse orthotopic xenograft model of nasopharyngeal carcinoma. 建立鼻咽癌可视化小鼠正位异种移植模型。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-08-29 DOI: 10.1080/15384047.2024.2382531
Wei Chen, Wei-Min Chen, Si-Xia Chen, Li Jiang, Ge-Ge Shu, Yuan-Xiu Yin, Zhi-Peng Quan, Zi-Yan Zhou, Ming-Jun Shen, Ya-Ting Qin, Chao-Lin Yang, Xue-Jin Su, Min Kang

Mouse orthotopic xenograft tumor models are commonly employed in studies investigating the mechanisms underlying the development and progression of tumors and their preclinical treatment. However, the unavailability of mature and visualized orthotopic xenograft models of nasopharyngeal carcinoma limits the development of treatment strategies for this cancer. The aim of this study was to provide a simple and reliable method for building an orthotopic xenograft model of nasopharyngeal carcinoma. Human nasopharyngeal carcinoma (C666-1-luc) cells, stably expressing the firefly luciferase gene, were injected subcutaneously into the right axilla of BALB/C nude mice. Four weeks later, the resulting subcutaneous tumors were cut into small blocks and grafted into the nasopharynx of immunodeficient BALB/C nude mice to induce tumor formation. Tumor growth was monitored by bioluminescence imaging and small animal magnetic resonance imaging (MRI). The expression of histological and immunological antigens associated with orthotopic xenograft nasopharyngeal carcinoma was analyzed by tissue section analysis and immunohistochemistry (IHC). A visualized orthotopic xenograft nasopharyngeal carcinoma model was successfully developed in this study. Luminescence signal detection, micro-MRI, and hematoxylin and eosin staining revealed the successful growth of tumors in the nasopharynx of the nude mice. Moreover, IHC analysis detected cytokeratin (CK), CK5/6, P40, and P63 expression in the orthotopic tumors, consistent with the reported expression of these antigens in human nasopharyngeal tumors. This study established a reproducible, visual, and less lethal orthotopic xenograft model of nasopharyngeal carcinoma, providing a platform for preclinical research.

小鼠正位异种移植肿瘤模型通常用于研究肿瘤发生、发展和临床前治疗的机制。然而,由于缺乏成熟的可视化鼻咽癌异位移植模型,限制了该癌症治疗策略的开发。本研究旨在提供一种建立鼻咽癌异位移植模型的简单而可靠的方法。将稳定表达萤火虫荧光素酶基因的人鼻咽癌(C666-1-luc)细胞皮下注射到 BALB/C 裸鼠的右腋窝。四周后,将皮下肿瘤切成小块,移植到免疫缺陷 BALB/C 裸鼠的鼻咽部,诱导肿瘤形成。肿瘤生长情况通过生物发光成像和小动物磁共振成像(MRI)进行监测。通过组织切片分析和免疫组织化学(IHC)分析了与正位异种鼻咽癌相关的组织学和免疫学抗原的表达。本研究成功建立了可视化的正位异种移植鼻咽癌模型。发光信号检测、显微核磁共振成像以及苏木精和伊红染色显示肿瘤在裸鼠鼻咽部成功生长。此外,IHC 分析检测到细胞角蛋白(CK)、CK5/6、P40 和 P63 在原位肿瘤中的表达,这与报道的人类鼻咽肿瘤中这些抗原的表达一致。这项研究建立了一个可重现、可视且致死率较低的鼻咽癌异位移植模型,为临床前研究提供了一个平台。
{"title":"Establishment of a visualized mouse orthotopic xenograft model of nasopharyngeal carcinoma.","authors":"Wei Chen, Wei-Min Chen, Si-Xia Chen, Li Jiang, Ge-Ge Shu, Yuan-Xiu Yin, Zhi-Peng Quan, Zi-Yan Zhou, Ming-Jun Shen, Ya-Ting Qin, Chao-Lin Yang, Xue-Jin Su, Min Kang","doi":"10.1080/15384047.2024.2382531","DOIUrl":"10.1080/15384047.2024.2382531","url":null,"abstract":"<p><p>Mouse orthotopic xenograft tumor models are commonly employed in studies investigating the mechanisms underlying the development and progression of tumors and their preclinical treatment. However, the unavailability of mature and visualized orthotopic xenograft models of nasopharyngeal carcinoma limits the development of treatment strategies for this cancer. The aim of this study was to provide a simple and reliable method for building an orthotopic xenograft model of nasopharyngeal carcinoma. Human nasopharyngeal carcinoma (C666-1-luc) cells, stably expressing the firefly luciferase gene, were injected subcutaneously into the right axilla of BALB/C nude mice. Four weeks later, the resulting subcutaneous tumors were cut into small blocks and grafted into the nasopharynx of immunodeficient BALB/C nude mice to induce tumor formation. Tumor growth was monitored by bioluminescence imaging and small animal magnetic resonance imaging (MRI). The expression of histological and immunological antigens associated with orthotopic xenograft nasopharyngeal carcinoma was analyzed by tissue section analysis and immunohistochemistry (IHC). A visualized orthotopic xenograft nasopharyngeal carcinoma model was successfully developed in this study. Luminescence signal detection, micro-MRI, and hematoxylin and eosin staining revealed the successful growth of tumors in the nasopharynx of the nude mice. Moreover, IHC analysis detected cytokeratin (CK), CK5/6, P40, and P63 expression in the orthotopic tumors, consistent with the reported expression of these antigens in human nasopharyngeal tumors. This study established a reproducible, visual, and less lethal orthotopic xenograft model of nasopharyngeal carcinoma, providing a platform for preclinical research.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"25 1","pages":"2382531"},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11364074/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142104699","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
Cancer Biology & Therapy
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