首页 > 最新文献

Cancer Genomics & Proteomics最新文献

英文 中文
Characterization of the Genomic Landscape in HPV-positive Cervical and Head and Neck Squamous Cell Carcinomas by Whole Genome Next Generation Sequencing. 全基因组下一代测序对hpv阳性宫颈和头颈部鳞状细胞癌基因组景观的表征。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-03-01 DOI: 10.21873/cgp.20496
Jianlan Ren, Nian Ma, Tyler Seckar, Sheynaz Bassa, Nicola Zetola, Surbhi Grover, Zhi Wei, Erle Robertson

Background/aim: In this study, we provide a comprehensive characterization of HPV-positive primary cervical cancers (CC) and HPV-positive head and neck squamous cell carcinomas (HNSCC) through whole genome next-generation sequencing. Human papillomavirus (HPV) infection, recognized as a definitive human carcinogen, is increasingly acknowledged for its role in development of human cancers. HPV-driven cervical cancers are among the leading causes of cancer-related deaths worldwide, while HPV-driven head and neck cancers exhibit distinct biological and clinical characteristics. Recent data has provided convincing evidence that HPV-related cervical cancer, like HPV head and neck cancer also predict better outcomes, with viral integration patterns further predicting disease related outcomes.

Materials and methods: We designed an experimental study that encompasses four pairs of HPV-positive patient samples with controls, utilizing state-of-the-art Next Generation Sequencing (NGS) technology including whole genome sequencing, transcriptome sequencing and virus integration.

Results: Multiple mutated genes, including TTN, COL6A3, and FLNA, were identified shared between CC and HNSCC. Additionally, we observed a notable proportion of pathways affected by oncogenic alterations, particularly in the RTK-RAS and NOTCH pathways, in both CC and HNSCC. Furthermore, we discovered a shared down-regulation of the Hedgehog signaling pathway based on transcriptome expression analysis in KEGG. We also identified RUNX2 and TFPI as sites of virus integration, and upstream as well as downstream pathway modulators, and represent potential targets for therapeutic interventions.

Conclusion: Overall, this study showed a thorough comparison between CC and HNSCC from multiple aspects, including gene variations, oncogenic pathways, KEGG enrichment and virus integration sites. However, further studies, which involve larger patient cohorts should be undertaken to further support these findings.

背景/目的:在本研究中,我们通过下一代全基因组测序对hpv阳性的原发性宫颈癌(CC)和hpv阳性的头颈部鳞状细胞癌(HNSCC)进行了全面的表征。人乳头瘤病毒(HPV)感染被认为是一种明确的人类致癌物,它在人类癌症发展中的作用日益得到承认。hpv驱动的宫颈癌是全球癌症相关死亡的主要原因之一,而hpv驱动的头颈癌表现出独特的生物学和临床特征。最近的数据提供了令人信服的证据,表明HPV相关的宫颈癌,如HPV头颈癌,也可以预测更好的结果,病毒整合模式进一步预测疾病相关的结果。材料和方法:我们设计了一项实验研究,包括四对hpv阳性患者样本和对照组,利用最先进的下一代测序(NGS)技术,包括全基因组测序、转录组测序和病毒整合。结果:发现CC和HNSCC共有TTN、COL6A3、FLNA等多个突变基因。此外,我们在CC和HNSCC中观察到受致癌改变影响的显著比例的通路,特别是RTK-RAS和NOTCH通路。此外,基于转录组表达分析,我们在KEGG中发现了Hedgehog信号通路的共同下调。我们还发现RUNX2和TFPI是病毒整合的位点,也是上游和下游途径调节剂,代表了治疗干预的潜在靶点。结论:总体而言,本研究从基因变异、致癌途径、KEGG富集和病毒整合位点等多个方面对CC和HNSCC进行了全面比较。然而,需要进一步的研究,包括更大的患者队列,以进一步支持这些发现。
{"title":"Characterization of the Genomic Landscape in HPV-positive Cervical and Head and Neck Squamous Cell Carcinomas by Whole Genome Next Generation Sequencing.","authors":"Jianlan Ren, Nian Ma, Tyler Seckar, Sheynaz Bassa, Nicola Zetola, Surbhi Grover, Zhi Wei, Erle Robertson","doi":"10.21873/cgp.20496","DOIUrl":"10.21873/cgp.20496","url":null,"abstract":"<p><strong>Background/aim: </strong>In this study, we provide a comprehensive characterization of HPV-positive primary cervical cancers (CC) and HPV-positive head and neck squamous cell carcinomas (HNSCC) through whole genome next-generation sequencing. Human papillomavirus (HPV) infection, recognized as a definitive human carcinogen, is increasingly acknowledged for its role in development of human cancers. HPV-driven cervical cancers are among the leading causes of cancer-related deaths worldwide, while HPV-driven head and neck cancers exhibit distinct biological and clinical characteristics. Recent data has provided convincing evidence that HPV-related cervical cancer, like HPV head and neck cancer also predict better outcomes, with viral integration patterns further predicting disease related outcomes.</p><p><strong>Materials and methods: </strong>We designed an experimental study that encompasses four pairs of HPV-positive patient samples with controls, utilizing state-of-the-art Next Generation Sequencing (NGS) technology including whole genome sequencing, transcriptome sequencing and virus integration.</p><p><strong>Results: </strong>Multiple mutated genes, including TTN, COL6A3, and FLNA, were identified shared between CC and HNSCC. Additionally, we observed a notable proportion of pathways affected by oncogenic alterations, particularly in the RTK-RAS and NOTCH pathways, in both CC and HNSCC. Furthermore, we discovered a shared down-regulation of the Hedgehog signaling pathway based on transcriptome expression analysis in KEGG. We also identified RUNX2 and TFPI as sites of virus integration, and upstream as well as downstream pathway modulators, and represent potential targets for therapeutic interventions.</p><p><strong>Conclusion: </strong>Overall, this study showed a thorough comparison between CC and HNSCC from multiple aspects, including gene variations, oncogenic pathways, KEGG enrichment and virus integration sites. However, further studies, which involve larger patient cohorts should be undertaken to further support these findings.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 2","pages":"188-207"},"PeriodicalIF":2.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11880920/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490815","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
Accumulating Genetic Mutations from Primary to Secondary Biliary Tract Cancers: Analysis of Four Patients With Metachronous Biliary Tract Cancer Using Comprehensive Genomic Profiling. 从原发性到继发性胆道癌累积基因突变:利用综合基因组图谱分析4例异时性胆道癌患者。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-03-01 DOI: 10.21873/cgp.20505
Toshio Kokuryo, Yoshio Koike, Junpei Yamaguchi, Masaki Sunagawa, Taisuke Baba, Nobuyuki Watanabe, Shunsuke Onoe, Takashi Mizuno, Tomoki Ebata

Background/aim: Metachronous biliary tract cancer (BTC) is a rare occurrence after curative resection of primary BTC. The genetic alterations and pathogenesis associated with metachronous BTC remain poorly understood.

Patients and methods: We analyzed four patients with metachronous BTC who underwent resection at the Nagoya University Hospital between 2010 and 2024. Gene panel examination was performed on both primary and secondary tumors using next-generation sequencing.

Results: The median interval between resection of the primary tumor and diagnosis of metachronous BTC was 24 months. Genetic alterations were observed in all paired primary and metachronous carcinomas. The number of genetic mutations was higher in metachronous lesions than in primary lesions. CDKN2A and SMAD4 were the most frequently mutated genes in all metachronous lesions. Common genetic mutations between primary and metachronous lesions were confirmed in all four cases, suggesting a common clonal origin.

Conclusion: This study demonstrated that characteristic genetic alterations and their accumulation play important roles in metachronous BTC. This suggests that the increasing burden of gene mutations may play a crucial role in the carcinogenesis of metachronous BTC. Further investigation is required to validate these findings and elucidate the underlying molecular mechanisms.

背景/目的:异时性胆道癌(BTC)是原发性胆道癌根治性切除后发生的罕见肿瘤。与异时性BTC相关的遗传改变和发病机制尚不清楚。患者和方法:我们分析了2010年至2024年间在名古屋大学医院接受切除的4例异时性BTC患者。采用新一代测序技术对原发性和继发性肿瘤进行基因面板检查。结果:从原发肿瘤切除到诊断为异时性BTC的中位时间间隔为24个月。在所有配对原发性癌和异时性癌中均观察到遗传改变。异时性病变中基因突变的数量高于原发性病变。CDKN2A和SMAD4是所有异时性病变中最常见的突变基因。原发性和异时性病变之间的共同基因突变在所有四个病例中都得到证实,表明共同的克隆起源。结论:本研究表明特征性遗传改变及其积累在异时性BTC中起重要作用。这表明基因突变负担的增加可能在异时性BTC的癌变中起关键作用。需要进一步的研究来验证这些发现并阐明潜在的分子机制。
{"title":"Accumulating Genetic Mutations from Primary to Secondary Biliary Tract Cancers: Analysis of Four Patients With Metachronous Biliary Tract Cancer Using Comprehensive Genomic Profiling.","authors":"Toshio Kokuryo, Yoshio Koike, Junpei Yamaguchi, Masaki Sunagawa, Taisuke Baba, Nobuyuki Watanabe, Shunsuke Onoe, Takashi Mizuno, Tomoki Ebata","doi":"10.21873/cgp.20505","DOIUrl":"10.21873/cgp.20505","url":null,"abstract":"<p><strong>Background/aim: </strong>Metachronous biliary tract cancer (BTC) is a rare occurrence after curative resection of primary BTC. The genetic alterations and pathogenesis associated with metachronous BTC remain poorly understood.</p><p><strong>Patients and methods: </strong>We analyzed four patients with metachronous BTC who underwent resection at the Nagoya University Hospital between 2010 and 2024. Gene panel examination was performed on both primary and secondary tumors using next-generation sequencing.</p><p><strong>Results: </strong>The median interval between resection of the primary tumor and diagnosis of metachronous BTC was 24 months. Genetic alterations were observed in all paired primary and metachronous carcinomas. The number of genetic mutations was higher in metachronous lesions than in primary lesions. <i>CDKN2A</i> and <i>SMAD4</i> were the most frequently mutated genes in all metachronous lesions. Common genetic mutations between primary and metachronous lesions were confirmed in all four cases, suggesting a common clonal origin.</p><p><strong>Conclusion: </strong>This study demonstrated that characteristic genetic alterations and their accumulation play important roles in metachronous BTC. This suggests that the increasing burden of gene mutations may play a crucial role in the carcinogenesis of metachronous BTC. Further investigation is required to validate these findings and elucidate the underlying molecular mechanisms.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 2","pages":"346-353"},"PeriodicalIF":2.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11880928/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490814","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
Engineered Methioninase-expressing Tumor-targeting Salmonella typhimurium A1-R Inhibits Syngeneic-Cancer Mouse Models by Depleting Tumor Methionine. 表达肿瘤靶向鼠伤寒沙门菌A1-R的工程蛋氨酸酶通过消耗肿瘤蛋氨酸抑制同基因肿瘤小鼠模型。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-03-01 DOI: 10.21873/cgp.20499
Yutaro Kubota, Ming Zhao, Qinghong Han, Yusuke Aoki, Noriyuki Masaki, Koya Obara, Sei Morinaga, Kohei Mizuta, Motokazu Sato, Michael Bouvet, Koichi Kubota, Takuya Tsunoda, Robert M Hoffman

Background/aim: We previously developed Salmonella typhimurium A1-R, which selectively targets and kills tumors. In the present study, we established recombinant methioninase (rMETase)-producing Salmonella typhimurium A1-R (A1-R-rMETase), by transfer of the Pseudomonas putida methioninase gene, to target methionine addiction of syngeneic-cancer mouse models.

Materials and methods: A plasmid containing the Pseudomonas putida methioninase gene was extracted from METase-producing recombinant E. coli and inserted into Salmonella typhimurium A1-R using electroporation. Lewis Lung Carcinoma (LLC) cells (106) were injected subcutaneously in male C57BL/6 mice aged 4-6 weeks. We determined that 108 Salmonella typhimurium A1-R-rMETase administered iv was a safe dosage in C57BL/6 mice and was used for efficacy studies on LLC tumors in C57BL/6 mice. Tumor size was measured with calipers three times per week for 3 weeks. On day 22, tumor methionine levels were measured using HPLC in the control mice injected with phosphate-buffered saline (PBS) and the mice injected with Salmonella typhimurium A1-R-rMETase.

Results: The mean LLC tumor size of each group on day 22 was as follows: PBS control: 741.5 mm3; mice injected with Salmonella typhimurium A1-R: 566.3 mm3 (p=0.370); and mice injected with Salmonella typhimurium A1-R-rMETase: 198.8 mm3 (p=0.0003 vs control and p=0.0117 vs. Salmonella A1-R). The mice injected with Salmonella typhimurium A1-R-rMETase showed a significantly lower mean tumor methionine level than mice injected with PBS (5.9 nM/mg protein vs 11.1 nM/mg protein, p=0.0095). Salmonella typhimurium A1-R-rMETase grew continuously in the tumors but not in the liver or spleen.

Conclusion: Tumor-targeting Salmonella typhimurium A1-R engineered to express the Pseudomonas putida methioninase gene, inhibited LLC tumor growth in a syngeneic mouse model and reduced the methionine level in the tumor. Salmonella typhimurium A1-R-rMETase combines the tumor targeting and killing capability of Salmonella typhimurium A1-R plus rMETase which targets the methionine addiction of cancer.

背景/目的:我们之前开发了鼠伤寒沙门氏菌A1-R,它可以选择性地靶向和杀死肿瘤。本研究通过转移恶臭假单胞菌的蛋氨酸酶基因,建立了产生鼠伤寒沙门氏菌A1-R (A1-R-rMETase)的重组蛋氨酸酶(rMETase),以靶向同基因癌小鼠的蛋氨酸依赖模型。材料与方法:从产mease的重组大肠杆菌中提取含有恶臭假单胞菌蛋氨酸酶基因的质粒,电穿孔插入鼠伤寒沙门菌A1-R中。将Lewis Lung Carcinoma (LLC)细胞(106)注射于4-6周龄雄性C57BL/6小鼠皮下。我们确定108鼠伤寒沙门菌A1-R-rMETase iv给药C57BL/6小鼠是安全剂量,并用于C57BL/6小鼠LLC肿瘤的疗效研究。每周用卡尺测量肿瘤大小3次,持续3周。第22天,用高效液相色谱法测定注射磷酸缓冲盐水(PBS)的对照小鼠和注射鼠伤寒沙门菌A1-R-rMETase的小鼠的肿瘤蛋氨酸水平。结果:各组第22天LLC肿瘤平均大小如下:PBS对照组:741.5 mm3;鼠伤寒沙门菌A1-R注射小鼠:566.3 mm3 (p=0.370);鼠伤寒沙门菌A1-R- rmetase注射小鼠:198.8 mm3 (p=0.0003 vs对照组,p=0.0117 vs沙门氏菌A1-R)。注射鼠伤寒沙门菌A1-R-rMETase小鼠的平均肿瘤蛋氨酸水平显著低于注射PBS小鼠(5.9 nM/mg蛋白vs 11.1 nM/mg蛋白,p=0.0095)。鼠伤寒沙门菌A1-R-rMETase在肿瘤中持续生长,但在肝脏和脾脏中没有。结论:鼠伤寒沙门菌A1-R工程化表达恶臭假单胞菌蛋氨酸酶基因,可抑制LLC肿瘤的生长,降低肿瘤中蛋氨酸水平。鼠伤寒沙门菌A1-R-rMETase结合了鼠伤寒沙门菌A1-R和rMETase的肿瘤靶向和杀伤能力,靶向癌症的蛋氨酸依赖。
{"title":"Engineered Methioninase-expressing Tumor-targeting <i>Salmonella typhimurium</i> A1-R Inhibits Syngeneic-Cancer Mouse Models by Depleting Tumor Methionine.","authors":"Yutaro Kubota, Ming Zhao, Qinghong Han, Yusuke Aoki, Noriyuki Masaki, Koya Obara, Sei Morinaga, Kohei Mizuta, Motokazu Sato, Michael Bouvet, Koichi Kubota, Takuya Tsunoda, Robert M Hoffman","doi":"10.21873/cgp.20499","DOIUrl":"10.21873/cgp.20499","url":null,"abstract":"<p><strong>Background/aim: </strong>We previously developed <i>Salmonella typhimurium</i> A1-R, which selectively targets and kills tumors. In the present study, we established recombinant methioninase (rMETase)-producing <i>Salmonella typhimurium</i> A1-R (A1-R-rMETase), by transfer of the <i>Pseudomonas putida methioninase</i> gene, to target methionine addiction of syngeneic-cancer mouse models.</p><p><strong>Materials and methods: </strong>A plasmid containing the <i>Pseudomonas putida methioninase</i> gene was extracted from METase-producing recombinant <i>E. coli</i> and inserted into <i>Salmonella typhimurium</i> A1-R using electroporation. Lewis Lung Carcinoma (LLC) cells (10<sup>6</sup>) were injected subcutaneously in male C57BL/6 mice aged 4-6 weeks. We determined that 10<sup>8</sup> <i>Salmonella typhimurium</i> A1-R-rMETase administered iv was a safe dosage in C57BL/6 mice and was used for efficacy studies on LLC tumors in C57BL/6 mice. Tumor size was measured with calipers three times per week for 3 weeks. On day 22, tumor methionine levels were measured using HPLC in the control mice injected with phosphate-buffered saline (PBS) and the mice injected with <i>Salmonella typhimurium</i> A1-R-rMETase.</p><p><strong>Results: </strong>The mean LLC tumor size of each group on day 22 was as follows: PBS control: 741.5 mm<sup>3</sup>; mice injected with <i>Salmonella typhimurium</i> A1-R: 566.3 mm<sup>3</sup> (<i>p</i>=0.370); and mice injected with <i>Salmonella typhimurium</i> A1-R-rMETase: 198.8 mm<sup>3</sup> (<i>p</i>=0.0003 <i>vs</i> control and <i>p</i>=0.0117 <i>vs. Salmonella</i> A1-R). The mice injected with <i>Salmonella typhimurium</i> A1-R-rMETase showed a significantly lower mean tumor methionine level than mice injected with PBS (5.9 nM/mg protein <i>vs</i> 11.1 nM/mg protein, <i>p</i>=0.0095). <i>Salmonella typhimurium</i> A1-R-rMETase grew continuously in the tumors but not in the liver or spleen.</p><p><strong>Conclusion: </strong>Tumor-targeting <i>Salmonella typhimurium</i> A1-R engineered to express the <i>Pseudomonas putida methioninase</i> gene, inhibited LLC tumor growth in a syngeneic mouse model and reduced the methionine level in the tumor. <i>Salmonella typhimurium</i> A1-R-rMETase combines the tumor targeting and killing capability of <i>Salmonella typhimurium</i> A1-R plus rMETase which targets the methionine addiction of cancer.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 2","pages":"247-257"},"PeriodicalIF":2.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11880925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490819","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
Loss of E-cadherin Activates EGFR-MEK/ERK Signaling, Promoting Cervical Cancer Progression. e -钙粘蛋白缺失激活EGFR-MEK/ERK信号,促进宫颈癌进展
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-03-01 DOI: 10.21873/cgp.20501
Hee Yun, Gwan Hee Han, Daniel J Wee, Doo-Byung Chay, Joon-Yong Chung, Jae-Hoon Kim, Hanbyoul Cho

Background/aim: This study investigated the relationship between E-cadherin down-regulation and enhanced pERK1/2 signaling in cervical cancer, evaluated their combined prognostic impact, and explored potential therapeutic targets.

Materials and methods: We analyzed 188 cervical cancer specimens and 300 normal cervical tissue samples using tissue microarray and immunohistochemistry. Small interfering RNA transfection and western blotting were used to study molecular interactions in cervical cancer cell lines.

Results: We observed a significant inverse correlation between E-cadherin and pERK1/2 expression, as well as poor disease-free survival and overall survival. Additionally, molecular analysis indicated that E-cadherin silencing enhanced ERK signaling and promoted cancer cell proliferation.

Conclusion: The findings suggest that E-cadherin and pERK1/2 are crucial biomarkers for cervical cancer prognosis and their interaction provides a potential target for therapeutic interventions. Further studies are recommended to explore these pathways in the clinical setting.

背景/目的:本研究探讨宫颈癌E-cadherin下调与pERK1/2信号增强的关系,评估二者对预后的综合影响,探索潜在的治疗靶点。材料与方法:采用组织芯片技术和免疫组化技术对188例宫颈癌标本和300例正常宫颈组织标本进行分析。应用小干扰RNA转染和western blotting技术研究宫颈癌细胞系分子间相互作用。结果:我们观察到E-cadherin与pERK1/2表达呈显著负相关,无病生存期和总生存期也呈显著负相关。此外,分子分析表明,E-cadherin沉默可增强ERK信号传导,促进癌细胞增殖。结论:E-cadherin和pERK1/2是宫颈癌预后的重要生物标志物,它们的相互作用为治疗干预提供了潜在的靶点。建议在临床环境中进一步研究这些途径。
{"title":"Loss of E-cadherin Activates EGFR-MEK/ERK Signaling, Promoting Cervical Cancer Progression.","authors":"Hee Yun, Gwan Hee Han, Daniel J Wee, Doo-Byung Chay, Joon-Yong Chung, Jae-Hoon Kim, Hanbyoul Cho","doi":"10.21873/cgp.20501","DOIUrl":"10.21873/cgp.20501","url":null,"abstract":"<p><strong>Background/aim: </strong>This study investigated the relationship between E-cadherin down-regulation and enhanced pERK1/2 signaling in cervical cancer, evaluated their combined prognostic impact, and explored potential therapeutic targets.</p><p><strong>Materials and methods: </strong>We analyzed 188 cervical cancer specimens and 300 normal cervical tissue samples using tissue microarray and immunohistochemistry. Small interfering RNA transfection and western blotting were used to study molecular interactions in cervical cancer cell lines.</p><p><strong>Results: </strong>We observed a significant inverse correlation between E-cadherin and pERK1/2 expression, as well as poor disease-free survival and overall survival. Additionally, molecular analysis indicated that E-cadherin silencing enhanced ERK signaling and promoted cancer cell proliferation.</p><p><strong>Conclusion: </strong>The findings suggest that E-cadherin and pERK1/2 are crucial biomarkers for cervical cancer prognosis and their interaction provides a potential target for therapeutic interventions. Further studies are recommended to explore these pathways in the clinical setting.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 2","pages":"271-284"},"PeriodicalIF":2.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11880930/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490842","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
DEFA1, Primarily Expressed at the Invasive Tumor Front, Promotes OSCC Cell Invasion and Tumor Growth. DEFA1主要表达于侵袭性肿瘤前沿,促进OSCC细胞侵袭和肿瘤生长。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-03-01 DOI: 10.21873/cgp.20504
Hojin Jeong, Sang Woong Park, Young Sun Hwang

Background/aim: The tumor microenvironment greatly influences cancer occurrence, progression, and treatment resistance, making it a key target alongside cancer cells. In squamous cell carcinoma, the invasive front is crucial for studying invasion mechanisms driven by the surrounding microenvironment and for identifying biomarkers to diagnose and predict invasive cancer. In this study, we aimed to elucidate the regulation of cancer characteristics through the interactions between factors at the invasive tumor front and the surrounding tumor microenvironment.

Materials and methods: The invasive tumor front (ITF) and tumor center (TC) of collective cancer invasion were analyzed using microarray to compare gene expression. A stable cell line with depleted DEFA1 expression was established, and its effect on cancer growth was observed using a mouse tongue xenograft model. Invasive activity was assessed using Transwell assays. Gene profiling of cancer cells and analysis of secreted proteins interacting with U937 monocytic cells during co-culture were conducted using QuantSeq 3' mRNA sequencing and LC-MS/MS analysis.

Results: DEFA1 was overexpressed at the ITF of collective cancer invasion. YD10B cells with depleted DEFA1 expression exhibited significantly reduced invasiveness and tumor growth without changes in the cell cycle distribution. Co-culture with U937 cells significantly enhanced the invasiveness of YD10B cells, which was inhibited by anti-DEFA1 treatment. QuantSeq 3' mRNA sequencing and LC-MS/MS analyses confirmed that DEFA1 derived from U937 cells increased the invasiveness of YD10B cells. Recombinant DEFA1 (rDEFA1) significantly enhanced the invasiveness of YD10B cells via the JNK MAPK/NF-[Formula: see text]B signaling pathway, independent of changes in DEFA1 expression within YD10B cells.

Conclusion: DEFA1 is crucial for cancer invasion and growth, and monocyte-derived DEFA1 exacerbates these traits. This study highlights DEFA1's role in promoting invasion at the tumor front, where interactions with the microenvironment are active.

背景/目的:肿瘤微环境对肿瘤的发生、进展和治疗耐药有很大影响,与癌细胞一起成为关键靶点。在鳞状细胞癌中,浸润前沿对于研究由周围微环境驱动的浸润机制以及识别生物标志物以诊断和预测浸润性癌症至关重要。在本研究中,我们旨在通过侵袭性肿瘤前沿因素与肿瘤周围微环境之间的相互作用来阐明肿瘤特征的调控。材料与方法:采用微阵列技术对肿瘤集体侵袭的侵袭性肿瘤前沿(invasive tumor front, ITF)和肿瘤中心(tumor center, TC)进行基因表达比较分析。建立了DEFA1表达缺失的稳定细胞系,并使用小鼠舌异种移植模型观察其对肿瘤生长的影响。采用Transwell法评估侵袭性活动。采用QuantSeq 3’mRNA测序和LC-MS/MS分析,对共培养过程中癌细胞的基因谱和与U937单核细胞相互作用的分泌蛋白进行分析。结果:DEFA1在肿瘤集体侵袭的ITF处过表达。DEFA1表达缺失的YD10B细胞侵袭性和肿瘤生长明显降低,但细胞周期分布没有改变。与U937细胞共培养可显著增强YD10B细胞的侵袭性,抗defa1处理可抑制YD10B细胞的侵袭性。QuantSeq 3’mRNA测序和LC-MS/MS分析证实,来自U937细胞的DEFA1增加了YD10B细胞的侵袭性。重组DEFA1 (rDEFA1)通过JNK MAPK/NF- B信号通路显著增强YD10B细胞的侵袭性,不依赖于YD10B细胞内DEFA1表达的变化。结论:DEFA1对肿瘤的侵袭和生长至关重要,单核细胞来源的DEFA1加剧了这些特征。这项研究强调了DEFA1在促进肿瘤前沿侵袭中的作用,在那里与微环境的相互作用是活跃的。
{"title":"DEFA1, Primarily Expressed at the Invasive Tumor Front, Promotes OSCC Cell Invasion and Tumor Growth.","authors":"Hojin Jeong, Sang Woong Park, Young Sun Hwang","doi":"10.21873/cgp.20504","DOIUrl":"10.21873/cgp.20504","url":null,"abstract":"<p><strong>Background/aim: </strong>The tumor microenvironment greatly influences cancer occurrence, progression, and treatment resistance, making it a key target alongside cancer cells. In squamous cell carcinoma, the invasive front is crucial for studying invasion mechanisms driven by the surrounding microenvironment and for identifying biomarkers to diagnose and predict invasive cancer. In this study, we aimed to elucidate the regulation of cancer characteristics through the interactions between factors at the invasive tumor front and the surrounding tumor microenvironment.</p><p><strong>Materials and methods: </strong>The invasive tumor front (ITF) and tumor center (TC) of collective cancer invasion were analyzed using microarray to compare gene expression. A stable cell line with depleted DEFA1 expression was established, and its effect on cancer growth was observed using a mouse tongue xenograft model. Invasive activity was assessed using Transwell assays. Gene profiling of cancer cells and analysis of secreted proteins interacting with U937 monocytic cells during co-culture were conducted using QuantSeq 3' mRNA sequencing and LC-MS/MS analysis.</p><p><strong>Results: </strong>DEFA1 was overexpressed at the ITF of collective cancer invasion. YD10B cells with depleted DEFA1 expression exhibited significantly reduced invasiveness and tumor growth without changes in the cell cycle distribution. Co-culture with U937 cells significantly enhanced the invasiveness of YD10B cells, which was inhibited by anti-DEFA1 treatment. QuantSeq 3' mRNA sequencing and LC-MS/MS analyses confirmed that DEFA1 derived from U937 cells increased the invasiveness of YD10B cells. Recombinant DEFA1 (rDEFA1) significantly enhanced the invasiveness of YD10B cells <i>via</i> the JNK MAPK/NF-[Formula: see text]B signaling pathway, independent of changes in DEFA1 expression within YD10B cells.</p><p><strong>Conclusion: </strong>DEFA1 is crucial for cancer invasion and growth, and monocyte-derived DEFA1 exacerbates these traits. This study highlights DEFA1's role in promoting invasion at the tumor front, where interactions with the microenvironment are active.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 2","pages":"326-345"},"PeriodicalIF":2.6,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11880931/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143490818","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
Evaluation of HMGB1 Expression as a Clinical Biomarker for Cholangiocarcinoma. HMGB1表达作为胆管癌临床生物标志物的评价
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-01-01 DOI: 10.21873/cgp.20489
Supakan Amontailak, Attapol Titapun, Apinya Jusakul, Raynoo Thanan, Phongsaran Kimawaha, Wassana Jamnongkan, Malinee Thanee, Papitchaya Sirithawat, Songpol Haohan, Anchalee Techasen

Background/aim: Cholangiocarcinoma (CCA) is an epithelial malignancy that is most prevalent in Southeast Asia, particularly in the northeast of Thailand. Identifying and establishing specific biomarkers of CCA is crucial for ensuring accurate prognosis and enabling effective treatment. High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) molecule that can be released by dead or injured cells and is associated with tumor progression. This study aimed to investigate the expression levels of HMGB1 in CCA.

Materials and methods: The clinical significance of HMGB1 levels was assessed by examining their correlation with patients' clinicopathological data. A bioinformatics analysis was conducted to examine HMGB1 mRNA expression and perform survival analysis. The expression levels of 137 tissue cases were evaluated using the immunohistochemical technique, whereas the serum levels of 31 cases were evaluated using indirect ELISA.

Results: The GEPIA analysis demonstrated that HMGB1 exhibited elevated mRNA expression in CCA compared to the normal group. Immunohistochemical staining revealed that HMGB1 expression was primarily localized in the nucleus. High HMGB1 expression was observed in 57.6% of tissue samples, while low expression was detected in 42.4%. There was a significant positive correlation between high HMGB1 expression and the extrahepatic type of CCA as well as lymph node metastasis. The measurement of HMGB1 levels were assessed using indirect ELISA in 31 CCA serum samples, where 51.6% exhibited elevated concentrations of HMGB1. Elevated serum HMGB1 levels were significantly associated with advanced tumor stages and high levels of bilirubin levels.

Conclusion: HMGB1 in both tissue biopsies and blood serum shows potential as a predictive biomarker in CCA patients. These biomarkers could form the basis for facilitating more effective treatment planning.

背景/目的:胆管癌(CCA)是一种上皮性恶性肿瘤,常见于东南亚,尤其是泰国东北部。识别和建立特异性的CCA生物标志物对于确保准确的预后和有效的治疗至关重要。高迁移率组框1 (HMGB1)是一种损伤相关分子模式(DAMP)分子,可由死亡或损伤细胞释放,并与肿瘤进展有关。本研究旨在探讨HMGB1在CCA中的表达水平。材料与方法:通过与患者临床病理资料的相关性来评价HMGB1水平的临床意义。采用生物信息学方法检测HMGB1 mRNA表达并进行生存分析。采用免疫组化法检测137例组织的表达水平,采用间接ELISA法检测31例血清的表达水平。结果:GEPIA分析显示,与正常组相比,CCA中HMGB1 mRNA表达升高。免疫组化染色显示HMGB1主要表达于细胞核。HMGB1在57.6%的组织样品中高表达,42.4%的组织样品中低表达。HMGB1高表达与CCA肝外类型及淋巴结转移呈显著正相关。在31份CCA血清样本中,采用间接ELISA法评估HMGB1水平,其中51.6%显示HMGB1浓度升高。血清HMGB1水平升高与肿瘤晚期和胆红素水平升高显著相关。结论:HMGB1在CCA患者的组织活检和血清中均显示出作为预测生物标志物的潜力。这些生物标记物可以形成促进更有效的治疗计划的基础。
{"title":"Evaluation of HMGB1 Expression as a Clinical Biomarker for Cholangiocarcinoma.","authors":"Supakan Amontailak, Attapol Titapun, Apinya Jusakul, Raynoo Thanan, Phongsaran Kimawaha, Wassana Jamnongkan, Malinee Thanee, Papitchaya Sirithawat, Songpol Haohan, Anchalee Techasen","doi":"10.21873/cgp.20489","DOIUrl":"10.21873/cgp.20489","url":null,"abstract":"<p><strong>Background/aim: </strong>Cholangiocarcinoma (CCA) is an epithelial malignancy that is most prevalent in Southeast Asia, particularly in the northeast of Thailand. Identifying and establishing specific biomarkers of CCA is crucial for ensuring accurate prognosis and enabling effective treatment. High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) molecule that can be released by dead or injured cells and is associated with tumor progression. This study aimed to investigate the expression levels of HMGB1 in CCA.</p><p><strong>Materials and methods: </strong>The clinical significance of HMGB1 levels was assessed by examining their correlation with patients' clinicopathological data. A bioinformatics analysis was conducted to examine HMGB1 mRNA expression and perform survival analysis. The expression levels of 137 tissue cases were evaluated using the immunohistochemical technique, whereas the serum levels of 31 cases were evaluated using indirect ELISA.</p><p><strong>Results: </strong>The GEPIA analysis demonstrated that HMGB1 exhibited elevated mRNA expression in CCA compared to the normal group. Immunohistochemical staining revealed that HMGB1 expression was primarily localized in the nucleus. High HMGB1 expression was observed in 57.6% of tissue samples, while low expression was detected in 42.4%. There was a significant positive correlation between high HMGB1 expression and the extrahepatic type of CCA as well as lymph node metastasis. The measurement of HMGB1 levels were assessed using indirect ELISA in 31 CCA serum samples, where 51.6% exhibited elevated concentrations of HMGB1. Elevated serum HMGB1 levels were significantly associated with advanced tumor stages and high levels of bilirubin levels.</p><p><strong>Conclusion: </strong>HMGB1 in both tissue biopsies and blood serum shows potential as a predictive biomarker in CCA patients. These biomarkers could form the basis for facilitating more effective treatment planning.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 1","pages":"81-89"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696321/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142892343","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
Regulation and Function of CCL2 and N-Myc in Retinoic Acid-treated Neuroblastoma Cells. CCL2和N-Myc在维甲酸处理的神经母细胞瘤细胞中的调控和功能。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-01-01 DOI: 10.21873/cgp.20490
Nanke Murra, Nina Sophie Pommert, Berit Schmidt, Reema Sami Issa, Meike Kaehler, Henrike Bruckmueller, Vera Tim, Ingolf Cascorbi, Vicki Waetzig

Background/aim: Treatment with retinoic acid (RA) often promotes neuroblastoma differentiation and growth inhibition, including the suppression of the expression of the MYCN oncogene. However, RA also targets protumoral chemokines, such as CCL2, which may contribute to the development of resistance. The present study aimed to investigate the regulation and function of CCL2 and N-Myc in RA-treated neuroblastoma cells.

Materials and methods: In Kelly or SH-SY5Y cells, viability was quantified by cell fitness assays. Expression was analyzed using quantitative PCR and the regulation of proteins using enzyme-linked immunoabsorbent assays (ELISA) or western blots.

Results: In MYCN-amplified Kelly cells, endogenous CCL2 levels were significantly lower compared to MYCN non-amplified SH-SY5Y cells. Treatment with 5 μM RA increased CCL2 release in both cell lines, but reduced N-Myc levels and cell numbers in Kelly cells. Over-expression of MYCN enhanced viability in SH-SY5Y cells, but did not affect RA-induced CCL2 release, while supplementation of CCL2 in Kelly cells did not prevent RA-mediated growth reduction. Impaired N-Myc or CCL2 signaling reduced the survival of all RA-treated cells and inhibition of N-Myc also decreased CCL2 levels. However, attenuated survival signaling was not generally associated with reduced levels of N-Myc or CCL2. Co-application of RA and the growth factor receptor inhibitors cediranib or crizotinib decreased N-Myc levels only in Kelly cells, while CCL2 release was dependent on the cell type and stimulus.

Conclusion: CCL2 and N-Myc promote the viability of RA-treated cells, although the levels of these mediators were not consistently correlated with cellular outcomes, especially during apoptotic signaling.

背景/目的:维甲酸(RA)治疗经常促进神经母细胞瘤的分化和生长抑制,包括抑制MYCN癌基因的表达。然而,RA也针对原肿瘤趋化因子,如CCL2,这可能有助于耐药性的发展。本研究旨在探讨CCL2和N-Myc在ra治疗的神经母细胞瘤细胞中的调控和功能。材料和方法:在Kelly或SH-SY5Y细胞中,通过细胞适应度测定测定细胞活力。用定量PCR分析表达,用酶联免疫吸附试验(ELISA)或western blots分析蛋白调控。结果:在MYCN扩增的Kelly细胞中,内源性CCL2水平明显低于MYCN未扩增的SH-SY5Y细胞。5 μM RA处理增加了两种细胞系的CCL2释放,但降低了Kelly细胞的N-Myc水平和细胞数量。MYCN的过表达增强了SH-SY5Y细胞的活力,但不影响ra诱导的CCL2释放,而在Kelly细胞中补充CCL2并不能阻止ra介导的生长减少。受损的N-Myc或CCL2信号降低了所有ra处理细胞的存活,N-Myc的抑制也降低了CCL2水平。然而,减弱的生存信号通常与N-Myc或CCL2水平的降低无关。RA与生长因子受体抑制剂cediranib或crizotinib共同应用仅在Kelly细胞中降低N-Myc水平,而CCL2的释放依赖于细胞类型和刺激。结论:CCL2和N-Myc促进ra处理细胞的活力,尽管这些介质的水平与细胞结局不一致,特别是在凋亡信号传导过程中。
{"title":"Regulation and Function of CCL2 and N-Myc in Retinoic Acid-treated Neuroblastoma Cells.","authors":"Nanke Murra, Nina Sophie Pommert, Berit Schmidt, Reema Sami Issa, Meike Kaehler, Henrike Bruckmueller, Vera Tim, Ingolf Cascorbi, Vicki Waetzig","doi":"10.21873/cgp.20490","DOIUrl":"10.21873/cgp.20490","url":null,"abstract":"<p><strong>Background/aim: </strong>Treatment with retinoic acid (RA) often promotes neuroblastoma differentiation and growth inhibition, including the suppression of the expression of the MYCN oncogene. However, RA also targets protumoral chemokines, such as CCL2, which may contribute to the development of resistance. The present study aimed to investigate the regulation and function of CCL2 and N-Myc in RA-treated neuroblastoma cells.</p><p><strong>Materials and methods: </strong>In Kelly or SH-SY5Y cells, viability was quantified by cell fitness assays. Expression was analyzed using quantitative PCR and the regulation of proteins using enzyme-linked immunoabsorbent assays (ELISA) or western blots.</p><p><strong>Results: </strong>In MYCN-amplified Kelly cells, endogenous CCL2 levels were significantly lower compared to MYCN non-amplified SH-SY5Y cells. Treatment with 5 μM RA increased CCL2 release in both cell lines, but reduced N-Myc levels and cell numbers in Kelly cells. Over-expression of MYCN enhanced viability in SH-SY5Y cells, but did not affect RA-induced CCL2 release, while supplementation of CCL2 in Kelly cells did not prevent RA-mediated growth reduction. Impaired N-Myc or CCL2 signaling reduced the survival of all RA-treated cells and inhibition of N-Myc also decreased CCL2 levels. However, attenuated survival signaling was not generally associated with reduced levels of N-Myc or CCL2. Co-application of RA and the growth factor receptor inhibitors cediranib or crizotinib decreased N-Myc levels only in Kelly cells, while CCL2 release was dependent on the cell type and stimulus.</p><p><strong>Conclusion: </strong>CCL2 and N-Myc promote the viability of RA-treated cells, although the levels of these mediators were not consistently correlated with cellular outcomes, especially during apoptotic signaling.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 1","pages":"90-102"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696317/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142892354","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
RFC3 Knockdown Decreases Cervical Cancer Cell Proliferation, Migration and Invasion. RFC3基因敲低可抑制宫颈癌细胞增殖、迁移和侵袭。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-01-01 DOI: 10.21873/cgp.20493
Jae Woong Koh, Seon-Joo Park

Background/aim: Replication factor C subunit 3 (RFC3) is a critical component of the replication factor C complex, which is essential for DNA replication and repair. Recent studies have highlighted the RFC3's significance in various cancer types. Herein, we aimed to elucidate its biological role in cervical cancer.

Materials and methods: Cervical cancer cells were transfected with RFC3 or control siRNA. Cell viability was assessed using the MTT assay over a 4-day period and its clonogenic potential was determined using colony formation assays. Flow cytometry analysis was performed to evaluate cell cycle distribution. Transwell migration and invasion assays were performed to assess the migration and invasion abilities of cervical cancer cells.

Results: RFC3 knockdown significantly inhibited cell proliferation, induced cell-cycle arrest, and decreased migration and invasion in HeLa and ME-180 cells compared to control siRNA-transfected cells.

Conclusion: The crucial role of RFC3 in cervical cancer progression is highlighted. RFC3 knockdown resulted in decreased cervical cancer cell proliferation, migration and invasion, suggesting its potential as a therapeutic target in cervical cancer.

背景/目的:复制因子C亚基3 (RFC3)是复制因子C复合体的重要组成部分,对DNA的复制和修复至关重要。最近的研究强调了RFC3在各种癌症类型中的重要性。在此,我们旨在阐明其在宫颈癌中的生物学作用。材料和方法:用RFC3或对照siRNA转染宫颈癌细胞。在4天的时间里,用MTT法评估细胞活力,用集落形成法确定其克隆潜能。流式细胞术分析细胞周期分布。采用Transwell迁移和侵袭试验评价宫颈癌细胞的迁移和侵袭能力。结果:与对照sirna转染的细胞相比,RFC3敲低显著抑制HeLa和ME-180细胞的增殖,诱导细胞周期阻滞,减少迁移和侵袭。结论:RFC3在宫颈癌进展中的重要作用得到强调。RFC3敲低可降低宫颈癌细胞的增殖、迁移和侵袭,提示其有可能成为宫颈癌的治疗靶点。
{"title":"RFC3 Knockdown Decreases Cervical Cancer Cell Proliferation, Migration and Invasion.","authors":"Jae Woong Koh, Seon-Joo Park","doi":"10.21873/cgp.20493","DOIUrl":"10.21873/cgp.20493","url":null,"abstract":"<p><strong>Background/aim: </strong>Replication factor C subunit 3 (RFC3) is a critical component of the replication factor C complex, which is essential for DNA replication and repair. Recent studies have highlighted the RFC3's significance in various cancer types. Herein, we aimed to elucidate its biological role in cervical cancer.</p><p><strong>Materials and methods: </strong>Cervical cancer cells were transfected with RFC3 or control siRNA. Cell viability was assessed using the MTT assay over a 4-day period and its clonogenic potential was determined using colony formation assays. Flow cytometry analysis was performed to evaluate cell cycle distribution. Transwell migration and invasion assays were performed to assess the migration and invasion abilities of cervical cancer cells.</p><p><strong>Results: </strong>RFC3 knockdown significantly inhibited cell proliferation, induced cell-cycle arrest, and decreased migration and invasion in HeLa and ME-180 cells compared to control siRNA-transfected cells.</p><p><strong>Conclusion: </strong>The crucial role of RFC3 in cervical cancer progression is highlighted. RFC3 knockdown resulted in decreased cervical cancer cell proliferation, migration and invasion, suggesting its potential as a therapeutic target in cervical cancer.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 1","pages":"127-135"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696326/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142892357","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
FN1 and VEGFA Are Potential Therapeutic Targets in Glioblastoma as Determined by Bioinformatics Analysis. 生物信息学分析确定FN1和VEGFA是胶质母细胞瘤的潜在治疗靶点。
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-01-01 DOI: 10.21873/cgp.20488
Mijung Im, Jungwook Roh, Wonyi Jang, Wanyeon Kim

Background/aim: Glioblastoma is the most malignant brain tumor, and despite advances in treatment, survival rates are still dismal. Therefore, a comprehensive understanding of the underlying molecular mechanisms of glioblastoma is needed. This study suggests potential therapeutic targets in glioblastoma that may provide new therapeutic insights.

Materials and methods: To identify hub genes in glioblastoma, three datasets were selected from the GEO database. After screening DEGs using GEO2R, GO and KEGG analyses were performed using DAVID. The PPI network was visualized using Cytoscape and 7 hub genes were extracted. The prognostic potential of 7 hub genes was investigated using the Gliovis and GEPIA2 databases.

Results: In total, 176 up-regulated and 263 down-regulated genes were identified. From the PPI network, 7 hub genes were identified including CAMK2A, DLG4, SNAP25, SYT1, MYC, FN1, and VEGFA. Out of the 7 hub genes identified, FN1 and VEGFA have been associated with a poor prognosis in glioblastoma based on the survival analysis.

Conclusion: This study suggests that high levels of FN1 and VEGFA expression are associated with a poor prognosis in glioblastoma and that both genes are promising targets for glioblastoma therapy. Bioinformatics analysis of DEGs revealed putative targets that might reveal the molecular mechanisms underlying glioblastoma.

背景/目的:胶质母细胞瘤是恶性程度最高的脑肿瘤,尽管治疗取得了进展,但其生存率仍然很低。因此,有必要全面了解胶质母细胞瘤的潜在分子机制。这项研究提示了胶质母细胞瘤的潜在治疗靶点,可能提供新的治疗见解。材料和方法:为了鉴定胶质母细胞瘤中的中枢基因,我们从GEO数据库中选择了三个数据集。在使用GEO2R筛选deg后,使用DAVID进行GO和KEGG分析。利用Cytoscape可视化PPI网络,提取7个枢纽基因。使用Gliovis和GEPIA2数据库研究7个枢纽基因的预后潜力。结果:共鉴定出176个上调基因和263个下调基因。从PPI网络中,鉴定出7个枢纽基因,包括CAMK2A、DLG4、SNAP25、SYT1、MYC、FN1和VEGFA。在鉴定的7个枢纽基因中,根据生存分析,FN1和VEGFA与胶质母细胞瘤的不良预后相关。结论:本研究提示,高水平的FN1和VEGFA表达与胶质母细胞瘤的不良预后相关,这两个基因都是胶质母细胞瘤治疗的有希望的靶点。deg的生物信息学分析揭示了可能揭示胶质母细胞瘤分子机制的假定靶点。
{"title":"FN1 and VEGFA Are Potential Therapeutic Targets in Glioblastoma as Determined by Bioinformatics Analysis.","authors":"Mijung Im, Jungwook Roh, Wonyi Jang, Wanyeon Kim","doi":"10.21873/cgp.20488","DOIUrl":"10.21873/cgp.20488","url":null,"abstract":"<p><strong>Background/aim: </strong>Glioblastoma is the most malignant brain tumor, and despite advances in treatment, survival rates are still dismal. Therefore, a comprehensive understanding of the underlying molecular mechanisms of glioblastoma is needed. This study suggests potential therapeutic targets in glioblastoma that may provide new therapeutic insights.</p><p><strong>Materials and methods: </strong>To identify hub genes in glioblastoma, three datasets were selected from the GEO database. After screening DEGs using GEO2R, GO and KEGG analyses were performed using DAVID. The PPI network was visualized using Cytoscape and 7 hub genes were extracted. The prognostic potential of 7 hub genes was investigated using the Gliovis and GEPIA2 databases.</p><p><strong>Results: </strong>In total, 176 up-regulated and 263 down-regulated genes were identified. From the PPI network, 7 hub genes were identified including CAMK2A, DLG4, SNAP25, SYT1, MYC, FN1, and VEGFA. Out of the 7 hub genes identified, FN1 and VEGFA have been associated with a poor prognosis in glioblastoma based on the survival analysis.</p><p><strong>Conclusion: </strong>This study suggests that high levels of FN1 and VEGFA expression are associated with a poor prognosis in glioblastoma and that both genes are promising targets for glioblastoma therapy. Bioinformatics analysis of DEGs revealed putative targets that might reveal the molecular mechanisms underlying glioblastoma.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 1","pages":"70-80"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696323/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142892344","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
FAM32A Suppression Decreases 5-Fluorouracil-induced Apoptosis and Is Associated With Poor Prognosis in Gastric Cancer. FAM32A抑制可减少5-氟尿嘧啶诱导的胃癌细胞凋亡并与不良预后相关
IF 2.6 4区 医学 Q2 GENETICS & HEREDITY Pub Date : 2025-01-01 DOI: 10.21873/cgp.20487
Yuya Agatsuma, Dai Shimizu, Shinichi Umeda, Haruyoshi Tanaka, Norifumi Hattori, Masamichi Hayashi, Mitsuro Kanda, Chie Tanaka, Goro Nakayama, Michitaka Fujiwara, Yasuhiro Kodera

Background/aim: The development of new biomarkers to predict cancer patient prognosis is expected to aid in treatment selection, contributing to improved outcomes. In this study, we extracted a candidate gene associated with patient prognosis from a public database and investigated the molecular and biological functions and clinical significance of the gene in gastric cancer.

Materials and methods: We analyzed The Cancer Genome Atlas database and identified the family with sequence similarity 32 member a (FAM32A) as a candidate gene. We investigated the clinicopathological significance of FAM32A mRNA and protein expression in 300 and 176 gastric cancer patients respectively. We evaluated the molecular and biological functions by suppressing FAM32A expression in gastric cancer cell lines using small interfering RNA.

Results: In the polymerase chain reaction (PCR) cohort, low FAM32A expression group showed significantly shorter disease-specific survival (DSS) [hazard ratio (HR)=1.586; 95% confidence interval (95% CI)=1.056-2.382, p=0.026]. In the immunohistochemistry cohort, the FAM32A(-) group had significantly shorter overall survival (HR=1.703; 95% CI=1.050-2.764, p=0.031) and DSS (HR=2.123; 95% CI=1.185-3.804, p=0.011). Multivariate Cox hazard analysis revealed that FAM32A(-) was an independent adverse prognostic factor for DSS (p<0.001). AGS cell lines with FAM32A knockdown exhibited significant resistance to 5-fluorouracil (5-FU) and reduced apoptosis upon 5-FU administration. Gene set enrichment analysis indicated decreased gene expression related to the p53 signaling pathway in AGS cells with FAM32A knockdown that were treated with 5-FU.

Conclusion: FAM32A suppression decreases 5-FU-induced apoptosis. Low FAM32A expression is associated with a poor prognosis in gastric cancer, suggesting its potential as a biomarker.

背景/目的:开发新的生物标志物来预测癌症患者的预后,有望帮助治疗选择,有助于改善预后。在本研究中,我们从公共数据库中提取了一个与患者预后相关的候选基因,并研究了该基因在胃癌中的分子生物学功能和临床意义。材料和方法:通过对The Cancer Genome Atlas数据库的分析,确定了序列相似度为32 member a (FAM32A)的家族作为候选基因。我们分别在300例和176例胃癌患者中研究FAM32A mRNA和蛋白表达的临床病理意义。我们利用小干扰RNA抑制FAM32A在胃癌细胞系中的表达,以评估其分子生物学功能。结果:在聚合酶链反应(PCR)队列中,FAM32A低表达组患者的疾病特异性生存期(DSS)显著缩短[危险比(HR)=1.586;95%置信区间(95% CI)=1.056 ~ 2.382, p=0.026]。在免疫组化队列中,FAM32A(-)组总生存期显著缩短(HR=1.703;95% CI=1.050-2.764, p=0.031)和DSS (HR=2.123;95% CI=1.185 ~ 3.804, p=0.011)。多因素Cox风险分析显示FAM32A(-)是DSS的独立不良预后因素(p结论:抑制FAM32A可减少5- fu诱导的细胞凋亡。FAM32A低表达与胃癌预后不良相关,提示其作为生物标志物的潜力。
{"title":"<i>FAM32A</i> Suppression Decreases 5-Fluorouracil-induced Apoptosis and Is Associated With Poor Prognosis in Gastric Cancer.","authors":"Yuya Agatsuma, Dai Shimizu, Shinichi Umeda, Haruyoshi Tanaka, Norifumi Hattori, Masamichi Hayashi, Mitsuro Kanda, Chie Tanaka, Goro Nakayama, Michitaka Fujiwara, Yasuhiro Kodera","doi":"10.21873/cgp.20487","DOIUrl":"10.21873/cgp.20487","url":null,"abstract":"<p><strong>Background/aim: </strong>The development of new biomarkers to predict cancer patient prognosis is expected to aid in treatment selection, contributing to improved outcomes. In this study, we extracted a candidate gene associated with patient prognosis from a public database and investigated the molecular and biological functions and clinical significance of the gene in gastric cancer.</p><p><strong>Materials and methods: </strong>We analyzed The Cancer Genome Atlas database and identified the family with sequence similarity 32 member a (FAM32A) as a candidate gene. We investigated the clinicopathological significance of FAM32A mRNA and protein expression in 300 and 176 gastric cancer patients respectively. We evaluated the molecular and biological functions by suppressing FAM32A expression in gastric cancer cell lines using small interfering RNA.</p><p><strong>Results: </strong>In the polymerase chain reaction (PCR) cohort, low FAM32A expression group showed significantly shorter disease-specific survival (DSS) [hazard ratio (HR)=1.586; 95% confidence interval (95% CI)=1.056-2.382, p=0.026]. In the immunohistochemistry cohort, the FAM32A(-) group had significantly shorter overall survival (HR=1.703; 95% CI=1.050-2.764, p=0.031) and DSS (HR=2.123; 95% CI=1.185-3.804, p=0.011). Multivariate Cox hazard analysis revealed that FAM32A(-) was an independent adverse prognostic factor for DSS (p<0.001). AGS cell lines with FAM32A knockdown exhibited significant resistance to 5-fluorouracil (5-FU) and reduced apoptosis upon 5-FU administration. Gene set enrichment analysis indicated decreased gene expression related to the p53 signaling pathway in AGS cells with FAM32A knockdown that were treated with 5-FU.</p><p><strong>Conclusion: </strong>FAM32A suppression decreases 5-FU-induced apoptosis. Low FAM32A expression is associated with a poor prognosis in gastric cancer, suggesting its potential as a biomarker.</p>","PeriodicalId":9516,"journal":{"name":"Cancer Genomics & Proteomics","volume":"22 1","pages":"55-69"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11696319/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142892339","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 Genomics & Proteomics
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:604180095
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