首页 > 最新文献

Cancer Biology & Therapy最新文献

英文 中文
The facilitating effects of KRT80 on chemoresistance, lipogenesis, and invasion of esophageal cancer. KRT80 对食管癌化疗抗性、脂肪生成和侵袭的促进作用
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-01-19 DOI: 10.1080/15384047.2024.2302162
Wen-Jing Yun, Jun Li, Nan-Chang Yin, Cong-Yu Zhang, Zheng-Guo Cui, Li Zhang, Hua-Chuan Zheng

Keratin 80 (KRT80) is a filament protein that makes up one of the major structural fibers of epithelial cells, and involved in cell differentiation and epithelial barrier integrity. Here, KRT80 mRNA expression was found to be higher in esophageal cancer than normal epithelium by RT-PCR and bioinformatics analysis (p < .05), opposite to KRT80 methylation (p < .05). There was a negative relationship between promoter methylation and expression level of KRT80 gene in esophageal cancer (p < .05). KRT80 mRNA expression was positively correlated with the differentiation, infiltration of immune cells, and poor prognosis of esophageal cancer (p < .05). KRT80 mRNA expression was positively linked to no infiltration of immune cells, the short survival time of esophageal cancers (p < .05). The differential genes of KRT80 mRNA were involved in fat digestion and metabolism, peptidase inhibitor, and intermediate filament, desosome, keratinocyte differentiation, epidermis development, keratinization, ECM regulator, complement cascade, metabolism of vitamins and co-factor (p < .05). KRT-80-related genes were classified into endocytosis, cell adhesion molecule binding, cadherin binding, cell-cell junction, cell leading edge, epidermal cell differentiation and development, T cell differentiation and receptor complex, plasma membrane receptor complex, external side of plasma membrane, metabolism of amino acids and catabolism of small molecules, and so forth (p < .05). KRT80 knockdown suppressed anti-apoptosis, anti-pyroptosis, migration, invasion, chemoresistance, and lipogenesis in esophageal cancer cells (p < .05), while ACC1 and ACLY overexpression reversed the inhibitory effects of KRT80 on lipogenesis and chemoresistance. These findings indicated that up-regulated expression of KRT80 might be involved in esophageal carcinogenesis and subsequent progression, aggravate aggressive phenotypes, and induced chemoresistance by lipid droplet assembly and ACC1- and ACLY-mediated lipogenesis.

角蛋白 80 (KRT80) 是一种丝状蛋白,是上皮细胞的主要结构纤维之一,参与细胞分化和上皮屏障完整性。通过 RT-PCR 和生物信息学分析发现,食管癌患者的 KRT80 mRNA 表达量高于正常上皮细胞(p p p p p p p p p
{"title":"The facilitating effects of KRT80 on chemoresistance, lipogenesis, and invasion of esophageal cancer.","authors":"Wen-Jing Yun, Jun Li, Nan-Chang Yin, Cong-Yu Zhang, Zheng-Guo Cui, Li Zhang, Hua-Chuan Zheng","doi":"10.1080/15384047.2024.2302162","DOIUrl":"10.1080/15384047.2024.2302162","url":null,"abstract":"<p><p>Keratin 80 (KRT80) is a filament protein that makes up one of the major structural fibers of epithelial cells, and involved in cell differentiation and epithelial barrier integrity. Here, KRT80 mRNA expression was found to be higher in esophageal cancer than normal epithelium by RT-PCR and bioinformatics analysis (<i>p</i> < .05), opposite to KRT80 methylation (<i>p</i> < .05). There was a negative relationship between promoter methylation and expression level of KRT80 gene in esophageal cancer (<i>p</i> < .05). KRT80 mRNA expression was positively correlated with the differentiation, infiltration of immune cells, and poor prognosis of esophageal cancer (<i>p</i> < .05). KRT80 mRNA expression was positively linked to no infiltration of immune cells, the short survival time of esophageal cancers (<i>p</i> < .05). The differential genes of KRT80 mRNA were involved in fat digestion and metabolism, peptidase inhibitor, and intermediate filament, desosome, keratinocyte differentiation, epidermis development, keratinization, ECM regulator, complement cascade, metabolism of vitamins and co-factor (<i>p</i> < .05). KRT-80-related genes were classified into endocytosis, cell adhesion molecule binding, cadherin binding, cell-cell junction, cell leading edge, epidermal cell differentiation and development, T cell differentiation and receptor complex, plasma membrane receptor complex, external side of plasma membrane, metabolism of amino acids and catabolism of small molecules, and so forth (<i>p</i> < .05). KRT80 knockdown suppressed anti-apoptosis, anti-pyroptosis, migration, invasion, chemoresistance, and lipogenesis in esophageal cancer cells (<i>p</i> < .05), while ACC1 and ACLY overexpression reversed the inhibitory effects of KRT80 on lipogenesis and chemoresistance. These findings indicated that up-regulated expression of KRT80 might be involved in esophageal carcinogenesis and subsequent progression, aggravate aggressive phenotypes, and induced chemoresistance by lipid droplet assembly and ACC1- and ACLY-mediated lipogenesis.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10802210/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139501973","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
FASN contributes to ADM resistance of diffuse large B-cell lymphoma by inhibiting ferroptosis via nf-κB/STAT3/GPX4 axis. FASN通过nf-κB/STAT3/GPX4轴抑制铁凋亡,从而促进弥漫大B细胞淋巴瘤对ADM的耐药性。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-09-30 DOI: 10.1080/15384047.2024.2403197
Xing Zhong, Weiwei Zhang, Weiming Zhang, Nasha Yu, Wuping Li, Xiangxiang Song

Drug resistance is a critical impediment to efficient therapy of diffuse large B-cell lymphoma (DLBCL) patients. Recent studies have highlighted the association between ferroptosis and drug resistance that has been reported. Fatty acid synthase (FASN) is always related to a poor prognosis. In this study, we investigate the impact of FASN on drug resistance in DLBCL and explore its potential modulation of ferroptosis mechanisms. The clinical correlation of FASN mRNA expression was first analyzed to confirm the role of FASN on drug resistance in DLBCL based on the TCGA database. Next, the impact of FASN on ferroptosis was investigated in vitro and in vivo. Furthermore, a combination of RNA-seq, western blot, luciferase reporter, and ChIP experiments was employed to elucidate the underlying mechanism. The prognosis for patients with DLBCL was worse when FASN was highly expressed, particularly in those undergoing chemotherapy for Adriamycin (ADM). FASN promoted tumor growth and resistance of DLBCL to ADM, both in vitro and in vivo. It is noteworthy that this effect was achieved by inhibiting ferroptosis, since Fer-1 (a ferroptosis inhibitor) treatment significantly recovered the effects of silencing FASN on inhibiting ferroptosis, while Erastin (a ferroptosis inducer) treatment attenuated the impact of overexpressing FASN. Mechanistically, FASN activated NF-κB/STAT3 signaling pathway through phosphorylating the upstream IKKα and IκBα, and the activated STAT3 promoted GPX4 expression by directly binding to GPX4 promoter. FASN inhibits ferroptosis in DLBCL via NF-κB/STAT3/GPX4 signaling pathway, indicating its critical role in mediating ADM resistance of DLBCL.

耐药性是弥漫大 B 细胞淋巴瘤(DLBCL)患者有效治疗的关键障碍。最近的研究强调了已报道的铁蛋白沉积与耐药性之间的关联。脂肪酸合成酶(FASN)总是与不良预后有关。在本研究中,我们研究了FASN对DLBCL耐药性的影响,并探讨了其对铁变态反应机制的潜在调节作用。首先,基于TCGA数据库分析了FASN mRNA表达的临床相关性,以确认FASN在DLBCL耐药性中的作用。接下来,研究人员在体外和体内研究了FASN对铁变态反应的影响。此外,研究人员还结合RNA-seq、Western blot、荧光素酶报告和ChIP实验来阐明其潜在机制。当FASN高表达时,DLBCL患者的预后较差,尤其是接受阿霉素(ADM)化疗的患者。FASN 在体外和体内都促进了 DLBCL 的肿瘤生长和对 ADM 的耐药性。值得注意的是,这种作用是通过抑制铁凋亡实现的,因为Fer-1(一种铁凋亡抑制剂)治疗能显著恢复沉默FASN对抑制铁凋亡的作用,而Erastin(一种铁凋亡诱导剂)治疗能减轻过表达FASN的影响。机制上,FASN通过磷酸化上游的IKKα和IκBα激活NF-κB/STAT3信号通路,激活的STAT3通过直接结合GPX4启动子促进GPX4的表达。FASN通过NF-κB/STAT3/GPX4信号通路抑制DLBCL中的铁突变,表明其在介导DLBCL的ADM耐药性中起着关键作用。
{"title":"FASN contributes to ADM resistance of diffuse large B-cell lymphoma by inhibiting ferroptosis via nf-κB/STAT3/GPX4 axis.","authors":"Xing Zhong, Weiwei Zhang, Weiming Zhang, Nasha Yu, Wuping Li, Xiangxiang Song","doi":"10.1080/15384047.2024.2403197","DOIUrl":"10.1080/15384047.2024.2403197","url":null,"abstract":"<p><p>Drug resistance is a critical impediment to efficient therapy of diffuse large B-cell lymphoma (DLBCL) patients. Recent studies have highlighted the association between ferroptosis and drug resistance that has been reported. Fatty acid synthase (FASN) is always related to a poor prognosis. In this study, we investigate the impact of FASN on drug resistance in DLBCL and explore its potential modulation of ferroptosis mechanisms. The clinical correlation of FASN mRNA expression was first analyzed to confirm the role of FASN on drug resistance in DLBCL based on the TCGA database. Next, the impact of FASN on ferroptosis was investigated in vitro and in vivo. Furthermore, a combination of RNA-seq, western blot, luciferase reporter, and ChIP experiments was employed to elucidate the underlying mechanism. The prognosis for patients with DLBCL was worse when FASN was highly expressed, particularly in those undergoing chemotherapy for Adriamycin (ADM). FASN promoted tumor growth and resistance of DLBCL to ADM, both in vitro and in vivo. It is noteworthy that this effect was achieved by inhibiting ferroptosis, since Fer-1 (a ferroptosis inhibitor) treatment significantly recovered the effects of silencing FASN on inhibiting ferroptosis, while Erastin (a ferroptosis inducer) treatment attenuated the impact of overexpressing FASN. Mechanistically, FASN activated NF-κB/STAT3 signaling pathway through phosphorylating the upstream IKKα and IκBα, and the activated STAT3 promoted GPX4 expression by directly binding to GPX4 promoter. FASN inhibits ferroptosis in DLBCL via NF-κB/STAT3/GPX4 signaling pathway, indicating its critical role in mediating ADM resistance of DLBCL.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11445901/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142342216","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
Correction. 更正。
IF 3.6 4区 医学 Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-12-31 Epub Date: 2023-12-26 DOI: 10.1080/15384047.2024.2299054
{"title":"Correction.","authors":"","doi":"10.1080/15384047.2024.2299054","DOIUrl":"10.1080/15384047.2024.2299054","url":null,"abstract":"","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10761110/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139037364","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
Hypoxia promotes non-small cell lung cancer cell stemness, migration, and invasion via promoting glycolysis by lactylation of SOX9. 低氧可通过 SOX9 的乳化作用促进糖酵解,从而促进非小细胞肺癌细胞的干性、迁移和侵袭。
IF 3.6 4区 医学 Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-12-31 Epub Date: 2024-01-16 DOI: 10.1080/15384047.2024.2304161
Fei Yan, Yue Teng, Xiaoyou Li, Yuejiao Zhong, Chunyi Li, Feng Yan, Xia He

Background: Lung cancer is the deadliest form of malignancy and the most common subtype is non-small cell lung cancer (NSCLC). Hypoxia is a typical feature of solid tumor microenvironment. In the current study, we clarified the effects of hypoxia on stemness and metastasis and the molecular mechanism.

Methods: The biological functions were assessed using the sphere formation assay, Transwell assay, and XF96 extracellular flux analyzer. The protein levels were detected by western blot. The lactylation modification was assessed by western blot and immunoprecipitation. The role of SOX9 in vivo was explored using a xenografted tumor model.

Results: We observed that hypoxia promoted sphere formation, migration, invasion, glucose consumption, lactate production, glycolysis, and global lactylation. Inhibition of glycolysis suppressed cell stemness, migration, invasion, and lactylation. Moreover, hypoxia increased the levels of SOX9 and lactylation of SOX9, whereas inhibition of glycolysis reversed the increase. Additionally, knockdown of SOX9 abrogated the promotion of cell stemness, migration, and invasion. In tumor-bearing mice, overexpression of SOX9 promoted tumor growth, and inhibition of glycolysis suppressed tumor growth.

Conclusion: Hypoxia induced the lactylation of SOX9 to promote stemness, migration, and invasion via promoting glycolysis. The findings suggested that targeting hypoxia may be an effective way for NSCLC treatment and reveal a new mechanism of hypoxia in NSCLC.

背景:肺癌是最致命的恶性肿瘤,最常见的亚型是非小细胞肺癌(NSCLC)。缺氧是实体瘤微环境的典型特征。本研究阐明了缺氧对干细胞和转移的影响及其分子机制:方法:使用球形成试验、Transwell 试验和 XF96 细胞外通量分析仪评估生物功能。蛋白水平通过 Western 印迹检测。乳化修饰通过 Western 印迹和免疫沉淀进行评估。利用异种移植肿瘤模型探讨了SOX9在体内的作用:结果:我们观察到缺氧促进了球体形成、迁移、侵袭、葡萄糖消耗、乳酸产生、糖酵解和全乳化。抑制糖酵解可抑制细胞干性、迁移、侵袭和乳化。此外,缺氧会增加 SOX9 和 SOX9 乳化的水平,而抑制糖酵解会逆转这种增加。此外,敲除SOX9可抑制对细胞干性、迁移和侵袭的促进作用。在肿瘤小鼠中,过表达 SOX9 会促进肿瘤生长,而抑制糖酵解则会抑制肿瘤生长:结论:缺氧诱导SOX9乳化,通过促进糖酵解促进干性、迁移和侵袭。研究结果表明,以缺氧为靶点可能是治疗 NSCLC 的有效方法,并揭示了 NSCLC 中缺氧的新机制。
{"title":"Hypoxia promotes non-small cell lung cancer cell stemness, migration, and invasion via promoting glycolysis by lactylation of SOX9.","authors":"Fei Yan, Yue Teng, Xiaoyou Li, Yuejiao Zhong, Chunyi Li, Feng Yan, Xia He","doi":"10.1080/15384047.2024.2304161","DOIUrl":"10.1080/15384047.2024.2304161","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is the deadliest form of malignancy and the most common subtype is non-small cell lung cancer (NSCLC). Hypoxia is a typical feature of solid tumor microenvironment. In the current study, we clarified the effects of hypoxia on stemness and metastasis and the molecular mechanism.</p><p><strong>Methods: </strong>The biological functions were assessed using the sphere formation assay, Transwell assay, and XF96 extracellular flux analyzer. The protein levels were detected by western blot. The lactylation modification was assessed by western blot and immunoprecipitation. The role of SOX9 in vivo was explored using a xenografted tumor model.</p><p><strong>Results: </strong>We observed that hypoxia promoted sphere formation, migration, invasion, glucose consumption, lactate production, glycolysis, and global lactylation. Inhibition of glycolysis suppressed cell stemness, migration, invasion, and lactylation. Moreover, hypoxia increased the levels of SOX9 and lactylation of SOX9, whereas inhibition of glycolysis reversed the increase. Additionally, knockdown of SOX9 abrogated the promotion of cell stemness, migration, and invasion. In tumor-bearing mice, overexpression of SOX9 promoted tumor growth, and inhibition of glycolysis suppressed tumor growth.</p><p><strong>Conclusion: </strong>Hypoxia induced the lactylation of SOX9 to promote stemness, migration, and invasion via promoting glycolysis. The findings suggested that targeting hypoxia may be an effective way for NSCLC treatment and reveal a new mechanism of hypoxia in NSCLC.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10793688/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139472321","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
KLF7 enhances the invasion and migration of colorectal cancer cells via the miR-139-5p/TPD52 axis. KLF7 通过 miR-139-5p/TPD52 轴增强结直肠癌细胞的侵袭和迁移。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-08-03 DOI: 10.1080/15384047.2024.2385172
Juan Zhang, Zhihan Li, Jiaxu Han, Zhongtao Tian, Qingyu Meng, Wenbo Niu

In this study, we aimed to investigate the molecular mechanism of Krüppel-like factor 7 (KLF7) in colorectal cancer (CRC) cell invasion and migration. The expression pattern of KLF7 in CRC tissues and the correlation between KLF7 expression and clinical symptoms of CRC were analyzed. CRC cell lines were transfected with si-KLF7, followed by qRT-PCR or western blot detection of KLF7, miR-139-5p, and tumor protein D52 (TPD52) expression, cell counting kit-8 (CCK-8) assay to detect cell viability, and transwell detection of invasion and migration. Chromatin immunoprecipitation (ChIP) analyzed the enrichment KLF7 in the miR-139-5p promoter. The dual-luciferase reporter assay verified the binding relationship between KLF7 and miR-139-5p, and between miR-139-5p and TPD52. In the subcutaneous tumorigenesis experiment, tumor growth was observed and ki67-positive expression was detected. KLF7 is abundantly expressed in CRC cells KLF7 silencing inhibits CRC cell viability, invasion, and migration. KLF7 represses miR-139-5p expression by binding to the miR-139-5p promoter. miR-139-5p targets TPD52 expression. miR-13-5p inhibition or TPD52 overexpression partially counteracted the effect of KLF7 silencing in CRC cells. KLF7 silencing suppresses tumor growth in vivo. In conclusion, KLF7 suppresses miR-139-5p expression by binding to the miR-139-5p promoter, thereby upregulating TPD52 expression and enhancing CRC cell invasion and migration.

本研究旨在探讨Krüppel样因子7(KLF7)在结直肠癌(CRC)细胞侵袭和迁移中的分子机制。研究分析了 KLF7 在 CRC 组织中的表达模式以及 KLF7 表达与 CRC 临床症状的相关性。用 si-KLF7 转染 CRC 细胞系,然后用 qRT-PCR 或 western 印迹法检测 KLF7、miR-139-5p 和肿瘤蛋白 D52(TPD52)的表达,用细胞计数试剂盒-8(CCK-8)检测细胞活力,并用透孔器检测侵袭和迁移。染色质免疫沉淀(ChIP)分析了 KLF7 在 miR-139-5p 启动子中的富集情况。双荧光素酶报告实验验证了 KLF7 与 miR-139-5p 之间以及 miR-139-5p 与 TPD52 之间的结合关系。在皮下肿瘤发生实验中,观察到肿瘤生长,并检测到 ki67 阳性表达。KLF7 在 CRC 细胞中大量表达,KLF7 沉默可抑制 CRC 细胞的活力、侵袭和迁移。KLF7通过与miR-139-5p启动子结合抑制miR-139-5p的表达,miR-139-5p靶向TPD52的表达。沉默 KLF7 可抑制肿瘤在体内的生长。总之,KLF7通过与miR-139-5p启动子结合来抑制miR-139-5p的表达,从而上调TPD52的表达并增强CRC细胞的侵袭和迁移。
{"title":"KLF7 enhances the invasion and migration of colorectal cancer cells via the miR-139-5p/TPD52 axis.","authors":"Juan Zhang, Zhihan Li, Jiaxu Han, Zhongtao Tian, Qingyu Meng, Wenbo Niu","doi":"10.1080/15384047.2024.2385172","DOIUrl":"10.1080/15384047.2024.2385172","url":null,"abstract":"<p><p>In this study, we aimed to investigate the molecular mechanism of Krüppel-like factor 7 (KLF7) in colorectal cancer (CRC) cell invasion and migration. The expression pattern of KLF7 in CRC tissues and the correlation between KLF7 expression and clinical symptoms of CRC were analyzed. CRC cell lines were transfected with si-KLF7, followed by qRT-PCR or western blot detection of KLF7, miR-139-5p, and tumor protein D52 (TPD52) expression, cell counting kit-8 (CCK-8) assay to detect cell viability, and transwell detection of invasion and migration. Chromatin immunoprecipitation (ChIP) analyzed the enrichment KLF7 in the miR-139-5p promoter. The dual-luciferase reporter assay verified the binding relationship between KLF7 and miR-139-5p, and between miR-139-5p and TPD52. In the subcutaneous tumorigenesis experiment, tumor growth was observed and ki67-positive expression was detected. KLF7 is abundantly expressed in CRC cells KLF7 silencing inhibits CRC cell viability, invasion, and migration. KLF7 represses miR-139-5p expression by binding to the miR-139-5p promoter. miR-139-5p targets TPD52 expression. miR-13-5p inhibition or TPD52 overexpression partially counteracted the effect of KLF7 silencing in CRC cells. KLF7 silencing suppresses tumor growth <i>in vivo</i>. In conclusion, KLF7 suppresses miR-139-5p expression by binding to the miR-139-5p promoter, thereby upregulating TPD52 expression and enhancing CRC cell invasion and migration.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299624/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141888508","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 role of liver cancer stem cells in hepatocellular carcinoma metastasis. 肝癌干细胞在肝细胞癌转移中的作用。
IF 3.6 4区 医学 Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-12-31 Epub Date: 2024-02-23 DOI: 10.1080/15384047.2024.2321768
Qinghui Niu, Susu Ye, Liu Zhao, Yanzhi Qian, Fengchao Liu

Metastasis accounts for the vast majority of cancer deaths; however, this complex process has yet to be fully explained. To form metastases, cancer cells must undergo a series of steps, known as the "Metastatic cascade", each of which requires a specific functional transformation. Cancer stem cells (CSCs) play a vital role in tumor metastasis, but their dynamic behavior and regulatory mechanisms have not been fully elucidated. Based on the "Metastatic cascade" theory, this review summarizes the effect of liver CSCs on the metastatic biological programs that underlie the dissemination and metastatic growth of cancer cells. Liver CSCs have the capacity to initiate distant organ metastasis via EMT, and the microenvironment transformation that supports the ability of these cells to disseminate, evade immune surveillance, dormancy, and regenerate metastasis. Understanding the heterogeneity and traits of liver CSCs in these processes is critical for developing strategies to prevent and treat metastasis of advanced hepatocellular carcinoma (HCC).

转移是绝大多数癌症死亡的原因;然而,这一复杂的过程尚未得到充分解释。要形成转移,癌细胞必须经历一系列步骤,即所谓的 "转移级联",其中每个步骤都需要特定的功能转变。癌症干细胞(CSCs)在肿瘤转移中扮演着重要角色,但其动态行为和调控机制尚未完全阐明。基于 "转移级联 "理论,本综述总结了肝脏干细胞对转移生物程序的影响,这些程序是癌细胞扩散和转移生长的基础。肝脏造血干细胞有能力通过EMT启动远处器官转移,而微环境的转变则支持这些细胞的扩散、逃避免疫监视、休眠和再生转移的能力。了解肝脏间充质干细胞在这些过程中的异质性和特征对于制定预防和治疗晚期肝细胞癌(HCC)转移的策略至关重要。
{"title":"The role of liver cancer stem cells in hepatocellular carcinoma metastasis.","authors":"Qinghui Niu, Susu Ye, Liu Zhao, Yanzhi Qian, Fengchao Liu","doi":"10.1080/15384047.2024.2321768","DOIUrl":"10.1080/15384047.2024.2321768","url":null,"abstract":"<p><p>Metastasis accounts for the vast majority of cancer deaths; however, this complex process has yet to be fully explained. To form metastases, cancer cells must undergo a series of steps, known as the \"Metastatic cascade\", each of which requires a specific functional transformation. Cancer stem cells (CSCs) play a vital role in tumor metastasis, but their dynamic behavior and regulatory mechanisms have not been fully elucidated. Based on the \"Metastatic cascade\" theory, this review summarizes the effect of liver CSCs on the metastatic biological programs that underlie the dissemination and metastatic growth of cancer cells. Liver CSCs have the capacity to initiate distant organ metastasis via EMT, and the microenvironment transformation that supports the ability of these cells to disseminate, evade immune surveillance, dormancy, and regenerate metastasis. Understanding the heterogeneity and traits of liver CSCs in these processes is critical for developing strategies to prevent and treat metastasis of advanced hepatocellular carcinoma (HCC).</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10896152/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139930205","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
Repression of the SUMO-conjugating enzyme UBC9 is associated with lowered double minutes and reduced tumor progression. SUMO-结合酶UBC9的抑制与双分钟降低和肿瘤进展减慢有关。
IF 3.6 4区 医学 Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-12-31 Epub Date: 2024-03-11 DOI: 10.1080/15384047.2024.2323768
Yusi Wang, Hongyan Zou, Wei Ji, Min Huang, Benhui You, Nan Sun, Yuandong Qiao, Peng Liu, Lidan Xu, Xuelong Zhang, Mengdi Cai, Ye Kuang, Songbin Fu, Wenjing Sun, Xueyuan Jia, Jie Wu

Double minutes (DMs), extrachromosomal gene fragments found within certain tumors, have been noted to carry onco- and drug resistance genes contributing to tumor pathogenesis and progression. After screening for SUMO-related molecule expression within various tumor sample and cell line databases, we found that SUMO-conjugating enzyme UBC9 has been associated with genome instability and tumor cell DM counts, which was confirmed both in vitro and in vivo. Karyotyping determined DM counts post-UBC9 knockdown or SUMOylation inhibitor 2-D08, while RT-qPCR and Western blot were used to measure DM-carried gene expression in vitro. In vivo, fluorescence in situ hybridization (FISH) identified micronucleus (MN) expulsion. Western blot and immunofluorescence staining were then used to determine DNA damage extent, and a reporter plasmid system was constructed to detect changes in homologous recombination (HR) and non-homologous end joining (NHEJ) pathways. Our research has shown that UBC9 inhibition is able to attenuate DM formation and lower DM-carried gene expression, in turn reducing tumor growth and malignant phenotype, via MN efflux of DMs and lowering NHEJ activity to increase DNA damage. These findings thus reveal a relationship between heightened UBC9 activity, increased DM counts, and tumor progression, providing a potential approach for targeted therapies, via UBC9 inhibition.

双分体(DMs)是在某些肿瘤内发现的染色体外基因片段,它携带的抗肿瘤和抗药性基因有助于肿瘤的发病和进展。在对各种肿瘤样本和细胞系数据库中的 SUMO 相关分子表达进行筛选后,我们发现 SUMO 结合酶 UBC9 与基因组不稳定性和肿瘤细胞 DM 数量有关,这一点在体外和体内都得到了证实。核型分析确定了UBC9敲除或SUMO化抑制剂2-D08后的DM数量,而RT-qPCR和Western印迹则用于测量体外DM携带基因的表达。在体内,荧光原位杂交(FISH)确定了微核(MN)的排出。然后用 Western 印迹和免疫荧光染色来确定 DNA 损伤程度,并构建了一个报告质粒系统来检测同源重组(HR)和非同源末端连接(NHEJ)途径的变化。我们的研究表明,抑制 UBC9 能够通过 DM 的 MN 外流和降低 NHEJ 活性来增加 DNA 损伤,从而减少 DM 的形成和降低携带 DM 的基因表达,进而减少肿瘤的生长和恶性表型。因此,这些发现揭示了 UBC9 活性增强、DM 数量增加和肿瘤进展之间的关系,为通过抑制 UBC9 进行靶向治疗提供了一种潜在的方法。
{"title":"Repression of the SUMO-conjugating enzyme UBC9 is associated with lowered double minutes and reduced tumor progression.","authors":"Yusi Wang, Hongyan Zou, Wei Ji, Min Huang, Benhui You, Nan Sun, Yuandong Qiao, Peng Liu, Lidan Xu, Xuelong Zhang, Mengdi Cai, Ye Kuang, Songbin Fu, Wenjing Sun, Xueyuan Jia, Jie Wu","doi":"10.1080/15384047.2024.2323768","DOIUrl":"10.1080/15384047.2024.2323768","url":null,"abstract":"<p><p>Double minutes (DMs), extrachromosomal gene fragments found within certain tumors, have been noted to carry onco- and drug resistance genes contributing to tumor pathogenesis and progression. After screening for SUMO-related molecule expression within various tumor sample and cell line databases, we found that SUMO-conjugating enzyme UBC9 has been associated with genome instability and tumor cell DM counts, which was confirmed both <i>in vitro</i> and <i>in vivo</i>. Karyotyping determined DM counts post-UBC9 knockdown or SUMOylation inhibitor 2-D08, while RT-qPCR and Western blot were used to measure DM-carried gene expression <i>in vitro</i>. <i>In vivo</i>, fluorescence in situ hybridization (FISH) identified micronucleus (MN) expulsion. Western blot and immunofluorescence staining were then used to determine DNA damage extent, and a reporter plasmid system was constructed to detect changes in homologous recombination (HR) and non-homologous end joining (NHEJ) pathways. Our research has shown that UBC9 inhibition is able to attenuate DM formation and lower DM-carried gene expression, in turn reducing tumor growth and malignant phenotype, via MN efflux of DMs and lowering NHEJ activity to increase DNA damage. These findings thus reveal a relationship between heightened UBC9 activity, increased DM counts, and tumor progression, providing a potential approach for targeted therapies, via UBC9 inhibition.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10936631/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140093447","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
CPT1A mediates the succinylation of SP5 which activates transcription of PDPK1 to promote the viability and glycolysis of prostate cancer cells. CPT1A 介导 SP5 的琥珀酰化,SP5 激活 PDPK1 的转录,从而促进前列腺癌细胞的活力和糖酵解。
IF 3.6 4区 医学 Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-12-31 Epub Date: 2024-03-17 DOI: 10.1080/15384047.2024.2329372
Shufeng Liu, Xiaoguang Chen, Liqi Zhang, Bo Lu

Succinylation modification involves in the progression of human cancers. The present study aimed to investigate the role of CPT1A, which is a succinyltransferase in the progression of prostate cancer (PCa). CCK-8 was used to detect the cell viability. Seahorse was performed to evaluate the cell glycolysis. Luciferase assay was used to detect the transcriptional regulation. ChIP was performed to assess the binding between transcriptional factors with the promoters. Co-IP was used to assess the binding between proteins. We found that CPT1A was highly expressed in PCa tissues and cell lines. Silencing of CPT1A inhibited the viability and glycolysis of PCa cells. Mechanistically, CPT1A promoted the succinylation of SP5, which strengthened the binding between SP5 and the promoter of PDPK1. SP5 activated PDPK1 transcription and PDPK1 activated the AKT/mTOR signal pathway. These findings might provide novel targets for the diagnosis or therapy of prostate cancer.

琥珀酰化修饰涉及人类癌症的进展。本研究旨在探讨琥珀酰基转移酶 CPT1A 在前列腺癌(PCa)进展过程中的作用。CCK-8用于检测细胞活力。海马试验用于评估细胞糖酵解。荧光素酶测定用于检测转录调控。ChIP 用于评估转录因子与启动子之间的结合。Co-IP 用于评估蛋白质之间的结合。我们发现 CPT1A 在 PCa 组织和细胞系中高表达。沉默CPT1A可抑制PCa细胞的活力和糖酵解。从机制上讲,CPT1A 促进了 SP5 的琥珀酰化,从而加强了 SP5 与 PDPK1 启动子的结合。SP5 激活了 PDPK1 的转录,而 PDPK1 激活了 AKT/mTOR 信号通路。这些发现可能会为前列腺癌的诊断或治疗提供新的靶点。
{"title":"CPT1A mediates the succinylation of SP5 which activates transcription of PDPK1 to promote the viability and glycolysis of prostate cancer cells.","authors":"Shufeng Liu, Xiaoguang Chen, Liqi Zhang, Bo Lu","doi":"10.1080/15384047.2024.2329372","DOIUrl":"10.1080/15384047.2024.2329372","url":null,"abstract":"<p><p>Succinylation modification involves in the progression of human cancers. The present study aimed to investigate the role of CPT1A, which is a succinyltransferase in the progression of prostate cancer (PCa). CCK-8 was used to detect the cell viability. Seahorse was performed to evaluate the cell glycolysis. Luciferase assay was used to detect the transcriptional regulation. ChIP was performed to assess the binding between transcriptional factors with the promoters. Co-IP was used to assess the binding between proteins. We found that CPT1A was highly expressed in PCa tissues and cell lines. Silencing of CPT1A inhibited the viability and glycolysis of PCa cells. Mechanistically, CPT1A promoted the succinylation of SP5, which strengthened the binding between SP5 and the promoter of PDPK1. SP5 activated PDPK1 transcription and PDPK1 activated the AKT/mTOR signal pathway. These findings might provide novel targets for the diagnosis or therapy of prostate cancer.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10950282/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140142788","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
METTL14 decreases FTH1 mRNA stability via m6A methylation to promote sorafenib-induced ferroptosis of cervical cancer. METTL14通过m6A甲基化降低FTH1 mRNA的稳定性,从而促进索拉非尼诱导的宫颈癌铁变态反应。
IF 4.4 4区 医学 Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-12-31 Epub Date: 2024-05-13 DOI: 10.1080/15384047.2024.2349429
Lijie Li, Jie Zeng, Sili He, Yanfei Yang, Chen Wang

Cervical cancer (CC) is a prevalent malignancy among women worldwide. This study was designed to investigate the role of METTL14 in sorafenib-induced ferroptosis in CC. METTL14 expression and m6A methylation were determined in CC tissues, followed by analyzes correlating these factors with clinical features. Subsequently, METTL14 was knocked down in CC cell lines, and the effects on cell proliferation, mitochondrial morphology and ferroptosis were assessed using CCK-8, microscopy, and markers associated with ferroptosis, respectively. The regulatory relationship between METTL14 and FTH1 was verified using qRT-PCR and luciferase reporter assays. The functional significance of this interaction was further investigated both in vitro and in vivo by co-transfecting cells with overexpression vectors or shRNAs targeting METTL14 and FTH1 after sorafenib treatment. METTL14 expression and m6A methylation were significantly reduced in CC tissues, and lower METTL14 expression levels were associated with a poorer CC patients' prognosis. Notably, METTL14 expression increased during sorafenib-induced ferroptosis, and METTL14 knockdown attenuated the ferroptotic response induced by sorafenib in CC cells. FTH1 was identified as a direct target of METTL14, with METTL14 overexpression leading to increased m6A methylation of FTH1 mRNA, resulting in reduced stability and expression of FTH1 in CC. Furthermore, FTH1 overexpression or treatment with LY294002 partially counteracted the promotion of sorafenib-induced ferroptosis by METTL14. In vivo xenograft experiments demonstrated that inhibiting METTL14 reduced the anticancer effects of sorafenib, whereas suppression of FTH1 significantly enhanced sorafenib-induced ferroptosis and increased its anticancer efficacy. METTL14 reduces FTH1 mRNA stability through m6A methylation, thereby enhancing sorafenib-induced ferroptosis, which contributes to suppressing CC progression via the PI3K/Akt signaling pathway.

宫颈癌(CC)是全球妇女中普遍存在的恶性肿瘤。本研究旨在探讨METTL14在索拉非尼诱导的宫颈癌铁中毒中的作用。研究测定了CC组织中METTL14的表达和m6A甲基化,并分析了这些因素与临床特征的相关性。随后,在CC细胞系中敲除METTL14,并使用CCK-8、显微镜和与铁变态相关的标记物分别评估其对细胞增殖、线粒体形态和铁变态的影响。通过 qRT-PCR 和荧光素酶报告实验验证了 METTL14 和 FTH1 之间的调控关系。索拉非尼治疗后,通过过表达载体或靶向 METTL14 和 FTH1 的 shRNAs 共同转染细胞,进一步研究了这种相互作用在体外和体内的功能意义。在CC组织中,METTL14的表达和m6A甲基化明显降低,而较低的METTL14表达水平与CC患者较差的预后有关。值得注意的是,在索拉非尼诱导的铁变态反应过程中,METTL14的表达会增加,而METTL14的敲除会减弱索拉非尼在CC细胞中诱导的铁变态反应。FTH1被确定为METTL14的直接靶标,METTL14过表达会导致FTH1 mRNA的m6A甲基化增加,从而降低FTH1在CC中的稳定性和表达。此外,FTH1过表达或用LY294002治疗可部分抵消METTL14对索拉非尼诱导的铁变态反应的促进作用。体内异种移植实验表明,抑制METTL14会降低索拉非尼的抗癌效果,而抑制FTH1则会显著增强索拉非尼诱导的铁细胞沉降,提高其抗癌疗效。METTL14通过m6A甲基化降低了FTH1 mRNA的稳定性,从而增强了索拉非尼诱导的铁变态反应,这有助于通过PI3K/Akt信号通路抑制CC的进展。
{"title":"METTL14 decreases FTH1 mRNA stability via m6A methylation to promote sorafenib-induced ferroptosis of cervical cancer.","authors":"Lijie Li, Jie Zeng, Sili He, Yanfei Yang, Chen Wang","doi":"10.1080/15384047.2024.2349429","DOIUrl":"10.1080/15384047.2024.2349429","url":null,"abstract":"<p><p>Cervical cancer (CC) is a prevalent malignancy among women worldwide. This study was designed to investigate the role of METTL14 in sorafenib-induced ferroptosis in CC. METTL14 expression and m6A methylation were determined in CC tissues, followed by analyzes correlating these factors with clinical features. Subsequently, METTL14 was knocked down in CC cell lines, and the effects on cell proliferation, mitochondrial morphology and ferroptosis were assessed using CCK-8, microscopy, and markers associated with ferroptosis, respectively. The regulatory relationship between METTL14 and FTH1 was verified using qRT-PCR and luciferase reporter assays. The functional significance of this interaction was further investigated both <i>in vitro</i> and <i>in vivo</i> by co-transfecting cells with overexpression vectors or shRNAs targeting METTL14 and FTH1 after sorafenib treatment. METTL14 expression and m6A methylation were significantly reduced in CC tissues, and lower METTL14 expression levels were associated with a poorer CC patients' prognosis. Notably, METTL14 expression increased during sorafenib-induced ferroptosis, and METTL14 knockdown attenuated the ferroptotic response induced by sorafenib in CC cells. FTH1 was identified as a direct target of METTL14, with METTL14 overexpression leading to increased m6A methylation of FTH1 mRNA, resulting in reduced stability and expression of FTH1 in CC. Furthermore, FTH1 overexpression or treatment with LY294002 partially counteracted the promotion of sorafenib-induced ferroptosis by METTL14. <i>In vivo</i> xenograft experiments demonstrated that inhibiting METTL14 reduced the anticancer effects of sorafenib, whereas suppression of FTH1 significantly enhanced sorafenib-induced ferroptosis and increased its anticancer efficacy. METTL14 reduces FTH1 mRNA stability through m6A methylation, thereby enhancing sorafenib-induced ferroptosis, which contributes to suppressing CC progression via the PI3K/Akt signaling pathway.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11093024/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140911480","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
TP53AIP1 induce autophagy via the AKT/mTOR signaling pathway in the breast cancer cells. TP53AIP1 通过 AKT/mTOR 信号通路诱导乳腺癌细胞自噬。
IF 4.4 4区 医学 Q2 ONCOLOGY Pub Date : 2024-12-31 Epub Date: 2024-09-02 DOI: 10.1080/15384047.2024.2398297
Shutian Liu, Ting Xu, Xi Chen, Li Tang, Longjiang Li, Li Zhang, Yongqiang Yang, Jiayi Huang

Breast cancer ranks the first in the incidence of female cancer and is the most common cancer threatening the life and health of women worldwide.Tumor protein p53-regulated apoptosis-inducing protein 1 (TP53AIP1) is a pro-apoptotic gene downstream of p53. However, the role of TP53AIP1 in BC needs to be investigated. In vitro and in vivo experiments were conducted to assess the biological functions and associated mechanisms. Several bioinformatics analyses were made, CCK8 assay, wound healing, transwell assays, colony formation assay, EDU, flow cytometry, Immunofluorescence, qRT-PCR and Western-blotting were performed. In our study, we discovered that BC samples had low levels of TP53AIP1 expression, which correlated with a lower survival rate in BC patients. When TP53AIP1 was up-regulated, it caused a decrease in cell proliferation, migration, and invasion. It also induced epithelial-to-mesenchymal transition (EMT) and protective autophagy. Furthermore, the over-expression of TP53AIP1 suppressed tumor growth when tested in vivo. We also noticed that TP53AIP1 up-regulation resulted in decreased levels of phosphorylation in AKT and mTOR, suggesting a mechanistic role. In addition, we performed functional rescue experiments where the activation of AKT was able to counteract the impact of TP53AIP1 on the survival and autophagy in breast cancer cell lines. This suggests that TP53AIP1 acts as an oncogene by controlling the AKT/mTOR pathway. These findings reveal TP53AIP1 as a gene that suppresses tumor growth and triggers autophagy through the AKT/mTOR pathway in breast cancer cells. As a result, TP53AIP1 presents itself as a potential target for novel therapeutic approaches in treating breast cancer.

肿瘤蛋白 p53 调控凋亡诱导蛋白 1(TP53AIP1)是 p53 的下游促凋亡基因。然而,TP53AIP1在乳腺癌中的作用还有待研究。为了评估其生物学功能和相关机制,我们进行了体外和体内实验。我们进行了多项生物信息学分析、CCK8 试验、伤口愈合试验、Transwell 试验、集落形成试验、EDU、流式细胞术、免疫荧光、qRT-PCR 和 Western 印迹。我们在研究中发现,BC样本的TP53AIP1表达水平较低,这与BC患者的生存率较低有关。当 TP53AIP1 上调时,它会导致细胞增殖、迁移和侵袭的减少。它还能诱导上皮细胞向间质转化(EMT)和保护性自噬。此外,在体内测试时,过度表达 TP53AIP1 可抑制肿瘤生长。我们还注意到,TP53AIP1 的上调导致 AKT 和 mTOR 的磷酸化水平下降,这表明了其机制作用。此外,我们还进行了功能性挽救实验,发现 AKT 的激活能够抵消 TP53AIP1 对乳腺癌细胞株存活和自噬的影响。这表明 TP53AIP1 是通过控制 AKT/mTOR 通路来充当致癌基因的。这些发现揭示了 TP53AIP1 是一种通过 AKT/mTOR 通路抑制肿瘤生长和引发乳腺癌细胞自噬的基因。因此,TP53AIP1 成为治疗乳腺癌的新型疗法的潜在靶点。
{"title":"TP53AIP1 induce autophagy via the AKT/mTOR signaling pathway in the breast cancer cells.","authors":"Shutian Liu, Ting Xu, Xi Chen, Li Tang, Longjiang Li, Li Zhang, Yongqiang Yang, Jiayi Huang","doi":"10.1080/15384047.2024.2398297","DOIUrl":"10.1080/15384047.2024.2398297","url":null,"abstract":"<p><p>Breast cancer ranks the first in the incidence of female cancer and is the most common cancer threatening the life and health of women worldwide.Tumor protein p53-regulated apoptosis-inducing protein 1 (TP53AIP1) is a pro-apoptotic gene downstream of p53. However, the role of TP53AIP1 in BC needs to be investigated. In vitro and in vivo experiments were conducted to assess the biological functions and associated mechanisms. Several bioinformatics analyses were made, CCK8 assay, wound healing, transwell assays, colony formation assay, EDU, flow cytometry, Immunofluorescence, qRT-PCR and Western-blotting were performed. In our study, we discovered that BC samples had low levels of TP53AIP1 expression, which correlated with a lower survival rate in BC patients. When TP53AIP1 was up-regulated, it caused a decrease in cell proliferation, migration, and invasion. It also induced epithelial-to-mesenchymal transition (EMT) and protective autophagy. Furthermore, the over-expression of TP53AIP1 suppressed tumor growth when tested in vivo. We also noticed that TP53AIP1 up-regulation resulted in decreased levels of phosphorylation in AKT and mTOR, suggesting a mechanistic role. In addition, we performed functional rescue experiments where the activation of AKT was able to counteract the impact of TP53AIP1 on the survival and autophagy in breast cancer cell lines. This suggests that TP53AIP1 acts as an oncogene by controlling the AKT/mTOR pathway. These findings reveal TP53AIP1 as a gene that suppresses tumor growth and triggers autophagy through the AKT/mTOR pathway in breast cancer cells. As a result, TP53AIP1 presents itself as a potential target for novel therapeutic approaches in treating breast cancer.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11376407/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142119072","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