首页 > 最新文献

Oncology Research最新文献

英文 中文
lncRNA SNHG4 enhanced gastric cancer progression by modulating miR-409-3p/CREB1 axis. lncRNA SNHG4通过调节miR-409-3p/CREB1轴促进胃癌进展。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.042281
Zhouyang Cheng, Yuchen Hua, Yang Cao, Jun Qin

Objective: Gastric cancer (GC) is a globally common cancer characterized by high incidence and mortality worldwide. Advances in the molecular understanding of GC provide promising targets for GC diagnosis and therapy. Long non-coding RNAs (lncRNAs) and their downstream regulators are regarded to be implicated in the progression of multiple types of malignancies. Studies have shown that the lncRNA small nucleolar RNA host gene 4 (SNHG4) serves as a tumor promoter in various malignancies, while its function in GC has yet to be characterized. Therefore, our study aimed to explore the role and underlying mechanism of SNHG4 in GC.

Methods: We used qRT-PCR to analyze SNHG4 expression in GC tissues and cells. Kaplan-Meier analysis was used to assess the correlation between SNHG4 expression and the survival rate of GC patients. Cellular function experiments such as CCK-8, BrdU, colony formation, flow cytometry analysis, and transwell were performed to explore the effects of SNHG4 on GC cell proliferation, apoptosis, cell cycle, migration, and invasion. We also established xenograft mouse models to explore the effect of SNHG4 on GC tumor growth. Mechanically, dual luciferase reporter assay was used to verify the interaction between SNHG4 and miR-409-3p and between miR-409-3p and cAMP responsive element binding protein 1 (CREB1).

Results: The results indicated that SNHG4 was overexpressed in GC tissues and cell lines, and was linked with poor survival rate of GC patients. SNHG4 promoted GC cell proliferation, migration, and invasion while inhibiting cell apoptosis and cell cycle arrest in vitro. The in vivo experiment indicated that SNHG4 facilitated GC tumor growth. Furthermore, SNHG4 was demonstrated to bind to miR-409-3p. Moreover, CREB1 was directly targeted by miR-409-3p. Rescue assays demonstrated that miR-409-3p deficiency reversed the suppressive impact of SNHG4 knockdown on GC cell malignancy. Additionally, miR-409-3p was also revealed to inhibit GC cell proliferation, migration, and invasion by targeting CREB1.

Conclusion: In conclusion, we verified that the SNHG4 promoted GC growth and metastasis by binding to miR-409-3p to upregulate CREB1, which may deepen the understanding of the underlying mechanism in GC development.

目的:胃癌是一种全球常见的癌症,在世界范围内具有高发病率和高死亡率的特点。对胃癌的分子认识的进展为胃癌的诊断和治疗提供了有希望的靶点。长链非编码rna (lncRNAs)及其下游调控因子被认为与多种类型恶性肿瘤的进展有关。研究表明lncRNA小核仁RNA宿主基因4 (SNHG4)在多种恶性肿瘤中起肿瘤启动子的作用,但其在GC中的功能尚不明确。因此,我们的研究旨在探讨SNHG4在GC中的作用及其机制。方法:采用qRT-PCR方法分析SNHG4在GC组织和细胞中的表达。采用Kaplan-Meier分析评估SNHG4表达与胃癌患者生存率的相关性。通过CCK-8、BrdU、菌落形成、流式细胞术分析、transwell等细胞功能实验,探讨SNHG4对胃癌细胞增殖、凋亡、细胞周期、迁移和侵袭的影响。我们还建立了异种移植小鼠模型,探讨SNHG4对胃癌肿瘤生长的影响。机械上,双荧光素酶报告试验用于验证SNHG4与miR-409-3p之间以及miR-409-3p与cAMP响应元件结合蛋白1 (CREB1)之间的相互作用。结果:SNHG4在胃癌组织和细胞系中过表达,与胃癌患者的低生存率有关。SNHG4促进GC细胞增殖、迁移和侵袭,抑制细胞凋亡和细胞周期阻滞。体内实验表明,SNHG4促进GC肿瘤生长。此外,SNHG4被证明与miR-409-3p结合。此外,miR-409-3p直接靶向CREB1。挽救实验表明,miR-409-3p缺失逆转了SNHG4敲低对GC细胞恶性肿瘤的抑制作用。此外,miR-409-3p还被发现通过靶向CREB1抑制GC细胞的增殖、迁移和侵袭。结论:综上所述,我们验证了SNHG4通过结合miR-409-3p上调CREB1来促进胃癌的生长和转移,这可能会加深对胃癌发生的潜在机制的理解。
{"title":"lncRNA SNHG4 enhanced gastric cancer progression by modulating miR-409-3p/CREB1 axis.","authors":"Zhouyang Cheng, Yuchen Hua, Yang Cao, Jun Qin","doi":"10.32604/or.2024.042281","DOIUrl":"10.32604/or.2024.042281","url":null,"abstract":"<p><strong>Objective: </strong>Gastric cancer (GC) is a globally common cancer characterized by high incidence and mortality worldwide. Advances in the molecular understanding of GC provide promising targets for GC diagnosis and therapy. Long non-coding RNAs (lncRNAs) and their downstream regulators are regarded to be implicated in the progression of multiple types of malignancies. Studies have shown that the lncRNA small nucleolar RNA host gene 4 (SNHG4) serves as a tumor promoter in various malignancies, while its function in GC has yet to be characterized. Therefore, our study aimed to explore the role and underlying mechanism of SNHG4 in GC.</p><p><strong>Methods: </strong>We used qRT-PCR to analyze SNHG4 expression in GC tissues and cells. Kaplan-Meier analysis was used to assess the correlation between SNHG4 expression and the survival rate of GC patients. Cellular function experiments such as CCK-8, BrdU, colony formation, flow cytometry analysis, and transwell were performed to explore the effects of SNHG4 on GC cell proliferation, apoptosis, cell cycle, migration, and invasion. We also established xenograft mouse models to explore the effect of SNHG4 on GC tumor growth. Mechanically, dual luciferase reporter assay was used to verify the interaction between SNHG4 and miR-409-3p and between miR-409-3p and cAMP responsive element binding protein 1 (CREB1).</p><p><strong>Results: </strong>The results indicated that SNHG4 was overexpressed in GC tissues and cell lines, and was linked with poor survival rate of GC patients. SNHG4 promoted GC cell proliferation, migration, and invasion while inhibiting cell apoptosis and cell cycle arrest <i>in vitro</i>. The <i>in vivo</i> experiment indicated that SNHG4 facilitated GC tumor growth. Furthermore, SNHG4 was demonstrated to bind to miR-409-3p. Moreover, CREB1 was directly targeted by miR-409-3p. Rescue assays demonstrated that miR-409-3p deficiency reversed the suppressive impact of SNHG4 knockdown on GC cell malignancy. Additionally, miR-409-3p was also revealed to inhibit GC cell proliferation, migration, and invasion by targeting CREB1.</p><p><strong>Conclusion: </strong>In conclusion, we verified that the SNHG4 promoted GC growth and metastasis by binding to miR-409-3p to upregulate CREB1, which may deepen the understanding of the underlying mechanism in GC development.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"185-198"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671409/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903408","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
Combination of fruquintinib with venetoclax for the treatment of colorectal cancer. fruquininib联合venetoclax治疗结直肠癌。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050047
Wei Zhang, Weicheng Wang, Rui Wang, Xiao Han, Lijun Zhu, Wenjie Guo, Yanhong Gu

Background: As a novel blocker of vascular endothelial growth factor receptor (VEGFR), fruquintinib has been approved for treating colorectal cancer (CRC). However, its dosage and therapeutic efficacy are limited by its widespread adverse reactions. Venetoclax, recognized as the initial inhibitor of B-cell lymphoma protein 2 (BCL2), has shown potential in boosting the effectiveness of immunotherapy against CRC. This study investigated the efficacy and mechanisms of fruquintinib combined with venetoclax in treating CRC.

Methods and materials: We developed a colon cancer mouse model with the CT26 colon cell line to demonstrate fruquintinib and venetoclax's efficacy against tumors. Then we employed various techniques to evaluate different aspects of the experimental outcomes. Immunohistochemistry was used to detect cell proliferation and angiogenesis in tumor tissues. Western blot analysis was utilized to examine the occurrence of cell apoptosis, and flow cytometry to quantitate immune cells within the tumor tissues. Moreover, immunofluorescence was employed to measure cytokine levels.

Results: The strongest inhibition on tumor growth was achieved by the combination of fruquintinib with venetoclax, as opposed to individual drug use. Venetoclax was found to amplify the impact of fruquintinib, leading to decreased cancer cell proliferation, increased cancer cell apoptosis, lowered angiogenesis, better vascular structure normalization, and improved immune cell infiltration.

Conclusion: Our findings indicate that the addition of venetoclax enhances the impact of fruquintinib on vascular normalization and modulation of the tumor immune microenvironment. Our study presents the justification for utilizing the fruquintinib and venetoclax combination in treating CRC. Venetoclax holds promise in being assimilated into anticancer medications for CRC.

背景:作为一种新型血管内皮生长因子受体(VEGFR)阻滞剂,fruquininib已被批准用于治疗结直肠癌(CRC)。然而,其广泛的不良反应限制了其剂量和治疗效果。Venetoclax被认为是b细胞淋巴瘤蛋白2 (BCL2)的初始抑制剂,已显示出提高CRC免疫治疗有效性的潜力。本研究探讨了fruquininib联合venetoclax治疗结直肠癌的疗效及机制。方法与材料:利用CT26结肠癌细胞系建立结肠癌小鼠模型,验证fruquininib和venetoclax的抗肿瘤作用。然后,我们采用各种技术来评估实验结果的不同方面。免疫组化检测肿瘤组织细胞增殖和血管生成。Western blot检测细胞凋亡的发生,流式细胞术检测肿瘤组织内免疫细胞的数量。此外,免疫荧光法检测细胞因子水平。结果:与单独用药相比,fruquininib与venetoclax联合使用对肿瘤生长的抑制作用最强。研究发现Venetoclax可放大fruquininib的作用,导致癌细胞增殖减少,癌细胞凋亡增加,血管生成降低,血管结构正常化改善,免疫细胞浸润改善。结论:我们的研究结果表明,venetoclax的加入增强了fruquininib对血管正常化和肿瘤免疫微环境调节的影响。我们的研究提出了使用fruquininib和venetoclax联合治疗CRC的理由。Venetoclax有望被吸收为CRC的抗癌药物。
{"title":"Combination of fruquintinib with venetoclax for the treatment of colorectal cancer.","authors":"Wei Zhang, Weicheng Wang, Rui Wang, Xiao Han, Lijun Zhu, Wenjie Guo, Yanhong Gu","doi":"10.32604/or.2024.050047","DOIUrl":"10.32604/or.2024.050047","url":null,"abstract":"<p><strong>Background: </strong>As a novel blocker of vascular endothelial growth factor receptor (VEGFR), fruquintinib has been approved for treating colorectal cancer (CRC). However, its dosage and therapeutic efficacy are limited by its widespread adverse reactions. Venetoclax, recognized as the initial inhibitor of B-cell lymphoma protein 2 (BCL2), has shown potential in boosting the effectiveness of immunotherapy against CRC. This study investigated the efficacy and mechanisms of fruquintinib combined with venetoclax in treating CRC.</p><p><strong>Methods and materials: </strong>We developed a colon cancer mouse model with the CT26 colon cell line to demonstrate fruquintinib and venetoclax's efficacy against tumors. Then we employed various techniques to evaluate different aspects of the experimental outcomes. Immunohistochemistry was used to detect cell proliferation and angiogenesis in tumor tissues. Western blot analysis was utilized to examine the occurrence of cell apoptosis, and flow cytometry to quantitate immune cells within the tumor tissues. Moreover, immunofluorescence was employed to measure cytokine levels.</p><p><strong>Results: </strong>The strongest inhibition on tumor growth was achieved by the combination of fruquintinib with venetoclax, as opposed to individual drug use. Venetoclax was found to amplify the impact of fruquintinib, leading to decreased cancer cell proliferation, increased cancer cell apoptosis, lowered angiogenesis, better vascular structure normalization, and improved immune cell infiltration.</p><p><strong>Conclusion: </strong>Our findings indicate that the addition of venetoclax enhances the impact of fruquintinib on vascular normalization and modulation of the tumor immune microenvironment. Our study presents the justification for utilizing the fruquintinib and venetoclax combination in treating CRC. Venetoclax holds promise in being assimilated into anticancer medications for CRC.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"225-234"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671418/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903379","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
Genetic signatures of ERCC1 and ERCC2 expression, along with SNPs variants, unveil favorable prognosis in SCLC patients undergoing platinum-based chemotherapy. ERCC1和ERCC2表达的遗传特征,以及snp变异,揭示了接受铂类化疗的SCLC患者的良好预后。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050161
Enrico Caliman, Sara Fancelli, Federico Scolari, Adriano Pasqui, Clara Manneschi, Daniele Lavacchi, Francesca Mazzoni, Francesca Gensini, Valeria Pasini, Camilla Eva Comin, Luca Voltolini, Serena Pillozzi, Lorenzo Antonuzzo

Background: Platinum chemotherapy (CT) remains the backbone of systemic therapy for patients with small-cell lung cancer (SCLC). The nucleotide excision repair (NER) pathway plays a central role in the repair of the DNA damage exerted by platinum agents. Alteration in this repair mechanism may affect patients' survival.

Materials and methods: We conducted a retrospective analysis of data from 38 patients with extensive disease (ED)-SCLC who underwent platinum-CT at the Clinical Oncology Unit, Careggi University Hospital, Florence (Italy), from 2015 to 2020. mRNA expression analysis and single nucleotide polymorphism (SNP) characterization of three NER pathway genes-namely ERCC1, ERCC2, and ERCC5-were performed on patient tumor samples.

Results: Overall, elevated expression of ERCC genes was observed in SCLC patients compared to healthy controls. Patients with low ERCC1 and ERCC5 expression levels exhibited a better median progression-free survival (mPFS = 7.1 vs. 4.9 months, p = 0.39 for ERCC1 and mPFS = 6.9 vs. 4.8 months, p = 0.093 for ERCC5) and overall survival (mOS = 8.7 vs. 6.0 months, p = 0.4 for ERCC1 and mOS = 7.2 vs. 6.2 months, p = 0.13 for ERCC5). Genotyping analysis of five SNPs of ERCC genes showed a longer survival in patients harboring the wild-type genotype or the heterozygous variant of the ERCC1 rs11615 SNP (p = 0.24 for PFS and p = 0.14 for OS) and of the rs13181 and rs1799793 ERCC2 SNPs (p = 0.43 and p = 0.26 for PFS and p = 0.21 and p = 0.16 for OS, respectively) compared to patients with homozygous mutant genotypes.

Conclusions: The comprehensive analysis of ERCC gene expression and SNP variants appears to identify patients who derive greater survival benefits from platinum-CT.

背景:铂化疗(CT)仍然是小细胞肺癌(SCLC)患者全身治疗的支柱。核苷酸切除修复(NER)途径在铂类药物对DNA损伤的修复中起核心作用。这种修复机制的改变可能会影响患者的生存。材料和方法:我们对2015年至2020年在意大利佛罗伦萨Careggi大学医院临床肿瘤科接受铂ct检查的38例广泛疾病(ED)-SCLC患者的数据进行了回顾性分析。对患者肿瘤样本进行三种NER通路基因ERCC1、ERCC2和ercc5的mRNA表达分析和单核苷酸多态性(SNP)表征。结果:总体而言,与健康对照组相比,SCLC患者中ERCC基因的表达升高。ERCC1和ERCC5低表达水平的患者表现出更好的中位无进展生存期(mPFS = 7.1 vs. 4.9个月,ERCC1 = 0.39, mPFS = 6.9 vs. 4.8个月,ERCC5 = 0.093)和总生存期(mOS = 8.7 vs. 6.0个月,ERCC1 = 0.4, mOS = 7.2 vs. 6.2个月,ERCC5 = 0.13)。ERCC基因的5个SNP基因分型分析显示,携带ERCC1 rs11615 SNP的野生型或杂合变体(PFS为p = 0.24, OS为p = 0.14)以及rs13181和rs1799793 ERCC2 SNP (PFS为p = 0.43和p = 0.26, OS为p = 0.21和p = 0.16)的患者的生存期比纯合突变型患者更长。结论:ERCC基因表达和SNP变异的综合分析似乎确定了从铂ct中获得更大生存益处的患者。
{"title":"Genetic signatures of <i>ERCC1</i> and <i>ERCC2</i> expression, along with SNPs variants, unveil favorable prognosis in SCLC patients undergoing platinum-based chemotherapy.","authors":"Enrico Caliman, Sara Fancelli, Federico Scolari, Adriano Pasqui, Clara Manneschi, Daniele Lavacchi, Francesca Mazzoni, Francesca Gensini, Valeria Pasini, Camilla Eva Comin, Luca Voltolini, Serena Pillozzi, Lorenzo Antonuzzo","doi":"10.32604/or.2024.050161","DOIUrl":"10.32604/or.2024.050161","url":null,"abstract":"<p><strong>Background: </strong>Platinum chemotherapy (CT) remains the backbone of systemic therapy for patients with small-cell lung cancer (SCLC). The nucleotide excision repair (NER) pathway plays a central role in the repair of the DNA damage exerted by platinum agents. Alteration in this repair mechanism may affect patients' survival.</p><p><strong>Materials and methods: </strong>We conducted a retrospective analysis of data from 38 patients with extensive disease (ED)-SCLC who underwent platinum-CT at the Clinical Oncology Unit, Careggi University Hospital, Florence (Italy), from 2015 to 2020. mRNA expression analysis and single nucleotide polymorphism (SNP) characterization of three NER pathway genes-namely <i>ERCC1</i>, <i>ERCC2</i>, and <i>ERCC5</i>-were performed on patient tumor samples.</p><p><strong>Results: </strong>Overall, elevated expression of <i>ERCC</i> genes was observed in SCLC patients compared to healthy controls. Patients with low <i>ERCC1</i> and <i>ERCC5</i> expression levels exhibited a better median progression-free survival (mPFS = 7.1 <i>vs</i>. 4.9 months, <i>p</i> = 0.39 for <i>ERCC1</i> and mPFS = 6.9 <i>vs</i>. 4.8 months, <i>p</i> = 0.093 for <i>ERCC5</i>) and overall survival (mOS = 8.7 <i>vs</i>. 6.0 months, <i>p</i> = 0.4 for <i>ERCC1</i> and mOS = 7.2 <i>vs</i>. 6.2 months, <i>p</i> = 0.13 for <i>ERCC5</i>). Genotyping analysis of five SNPs of <i>ERCC</i> genes showed a longer survival in patients harboring the wild-type genotype or the heterozygous variant of the <i>ERCC1</i> rs11615 SNP (<i>p</i> = 0.24 for PFS and <i>p</i> = 0.14 for OS) and of the rs13181 and rs1799793 <i>ERCC2</i> SNPs (<i>p</i> = 0.43 and <i>p</i> = 0.26 for PFS and <i>p</i> = 0.21 and <i>p</i> = 0.16 for OS, respectively) compared to patients with homozygous mutant genotypes.</p><p><strong>Conclusions: </strong>The comprehensive analysis of <i>ERCC</i> gene expression and SNP variants appears to identify patients who derive greater survival benefits from platinum-CT.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"45-55"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671403/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903396","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
HNRNPC as a pan-cancer biomarker and therapeutic target involved in tumor progression and immune regulation. HNRNPC作为泛癌症生物标志物和治疗靶点参与肿瘤进展和免疫调节。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.055866
Yuezhou Zhang, Zhao Zhang, Jinxin Dong, Changan Liu

Background: Aberrant expression of RNA-binding proteins (RBPs) has been linked to a variety of diseases, including hematological disorders, cardiovascular diseases, and multiple types of cancer. Heterogeneous nuclear ribonucleoprotein C (HNRNPC), a member belonging to the heterogeneous nuclear ribonucleoprotein (hnRNP) family, plays a pivotal role in nucleic acid metabolism. Previous studies have underscored the significance of HNRNPC in tumorigenesis; however, its specific role in malignant tumor progression remains inadequately characterized.

Methods: We leveraged publicly available databases, including The Cancer Genome Atlas (TCGA), to explore the potential involvement of HNRNPC across various cancers. Additionally, we performed experimental validation studies focused on liver cancer.

Results: Our analysis revealed that HNRNPC is overexpressed in a wide range of common malignancies, including liver and lung cancers, and is strongly linked to unfavorable outcomes. Furthermore, HNRNPC was observed to be closely linked to tumor immunity. Through immune checkpoint analysis and immune cell infiltration assessment, HNRNPC emerged as a potential target for modulating tumor immunotherapy. Notably, silencing of HNRNPC markedly inhibited the proliferation, metastasis, and infiltration of liver cancer cells.

Conclusion: In summary, our findings highlight HNRNPC as a prognostic marker in various cancers, including liver cancer, and suggest its involvement in shaping the tumor immune microenvironment. These insights offer potential avenues for improving clinical outcomes in tumors with elevated HNRNPC expression, particularly through immunotherapeutic strategies.

背景:rna结合蛋白(rbp)的异常表达与多种疾病有关,包括血液系统疾病、心血管疾病和多种类型的癌症。异质核糖核蛋白C (HNRNPC)是异质核糖核蛋白(hnRNP)家族的一员,在核酸代谢中起着举足轻重的作用。先前的研究强调了HNRNPC在肿瘤发生中的重要性;然而,其在恶性肿瘤进展中的具体作用仍未得到充分的描述。方法:我们利用公开可用的数据库,包括癌症基因组图谱(TCGA),来探索HNRNPC在各种癌症中的潜在作用。此外,我们还进行了针对肝癌的实验验证研究。结果:我们的分析显示,HNRNPC在广泛的常见恶性肿瘤中过表达,包括肝癌和肺癌,并且与不良结果密切相关。此外,HNRNPC被观察到与肿瘤免疫密切相关。通过免疫检查点分析和免疫细胞浸润评估,HNRNPC成为调节肿瘤免疫治疗的潜在靶点。值得注意的是,沉默HNRNPC可显著抑制肝癌细胞的增殖、转移和浸润。结论:总之,我们的研究结果突出了HNRNPC作为包括肝癌在内的各种癌症的预后标志物,并提示其参与塑造肿瘤免疫微环境。这些见解为改善HNRNPC表达升高的肿瘤的临床结果提供了潜在的途径,特别是通过免疫治疗策略。
{"title":"HNRNPC as a pan-cancer biomarker and therapeutic target involved in tumor progression and immune regulation.","authors":"Yuezhou Zhang, Zhao Zhang, Jinxin Dong, Changan Liu","doi":"10.32604/or.2024.055866","DOIUrl":"10.32604/or.2024.055866","url":null,"abstract":"<p><strong>Background: </strong>Aberrant expression of RNA-binding proteins (RBPs) has been linked to a variety of diseases, including hematological disorders, cardiovascular diseases, and multiple types of cancer. Heterogeneous nuclear ribonucleoprotein C (HNRNPC), a member belonging to the heterogeneous nuclear ribonucleoprotein (hnRNP) family, plays a pivotal role in nucleic acid metabolism. Previous studies have underscored the significance of HNRNPC in tumorigenesis; however, its specific role in malignant tumor progression remains inadequately characterized.</p><p><strong>Methods: </strong>We leveraged publicly available databases, including The Cancer Genome Atlas (TCGA), to explore the potential involvement of HNRNPC across various cancers. Additionally, we performed experimental validation studies focused on liver cancer.</p><p><strong>Results: </strong>Our analysis revealed that HNRNPC is overexpressed in a wide range of common malignancies, including liver and lung cancers, and is strongly linked to unfavorable outcomes. Furthermore, HNRNPC was observed to be closely linked to tumor immunity. Through immune checkpoint analysis and immune cell infiltration assessment, HNRNPC emerged as a potential target for modulating tumor immunotherapy. Notably, silencing of HNRNPC markedly inhibited the proliferation, metastasis, and infiltration of liver cancer cells.</p><p><strong>Conclusion: </strong>In summary, our findings highlight HNRNPC as a prognostic marker in various cancers, including liver cancer, and suggest its involvement in shaping the tumor immune microenvironment. These insights offer potential avenues for improving clinical outcomes in tumors with elevated HNRNPC expression, particularly through immunotherapeutic strategies.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"83-102"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671622/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903398","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
Deep learning identification of novel autophagic protein-protein interactions and experimental validation of Beclin 2-Ubiquilin 1 axis in triple-negative breast cancer. 三阴性乳腺癌中新型自噬蛋白相互作用的深度学习鉴定和Beclin 2-Ubiquilin 1轴的实验验证。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.055921
Xiang Li, Wenke Jin, Lifeng Wu, Huan Wang, Xin Xie, Wei Huang, B O Liu

Background: Triple-negative breast cancer (TNBC), characterized by its lack of traditional hormone receptors and HER2, presents a significant challenge in oncology due to its poor response to conventional therapies. Autophagy is an important process for maintaining cellular homeostasis, and there are currently autophagy biomarkers that play an effective role in the clinical treatment of tumors. In contrast to targeting protein activity, intervention with protein-protein interaction (PPI) can avoid unrelated crosstalk and regulate the autophagy process with minimal interference pathways.

Methods: Here, we employed Naive Bayes, Decision Tree, and k-Nearest Neighbors to elucidate the complex PPI network associated with autophagy in TNBC, aiming to uncover novel therapeutic targets. Meanwhile, the candidate proteins interacting with Beclin 2 were initially screened in MDA-MB-231 cells using Beclin 2 as bait protein by immunoprecipitation-mass spectrometry assay, and the interaction relationship was verified by molecular docking and CO-IP experiments after intersection. Colony formation, cellular immunofluorescence, cell scratch and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) tests were used to predict the clinical therapeutic effects of manipulating candidate PPI.

Results: By developing three PPI classification models and analyzing over 13,000 datasets, we identified 3733 previously unknown autophagy-related PPIs. Our network analysis revealed the central role of Beclin 2 in autophagy regulation, uncovering its interactions with 39 newly identified proteins. Notably, the CO-IP studies identified the substantial interaction between Beclin 2 and Ubiquilin 1, which was anticipated by our model and discovered in immunoprecipitation-mass spectrometry assay results. Subsequently, in vitro investigations showed that overexpressing Beclin 2 increased Ubiquilin 1, promoted autophagy-dependent cell death, and inhibited proliferation and metastasis in MDA-MB-231 cells.

Conclusions: This study not only enhances our understanding of autophagy regulation in TNBC but also identifies the Beclin 2-Ubiquilin 1 axis as a promising target for precision therapy. These findings open new avenues for drug discovery and offer inspiration for more effective treatments for this aggressive cancer subtype.

背景:三阴性乳腺癌(TNBC)的特点是缺乏传统的激素受体和HER2,由于其对传统治疗的不良反应,在肿瘤学中提出了重大挑战。自噬是维持细胞稳态的重要过程,目前已有自噬生物标志物在肿瘤的临床治疗中发挥有效作用。与靶向蛋白活性相比,通过蛋白-蛋白相互作用(protein-protein interaction, PPI)干预可以避免不相关的串扰,以最小的干扰途径调节自噬过程。方法:本研究采用朴素贝叶斯、决策树和k近邻来阐明与TNBC自噬相关的复杂PPI网络,旨在发现新的治疗靶点。同时,以Beclin 2为诱饵蛋白,通过免疫沉淀-质谱法在MDA-MB-231细胞中初步筛选出与Beclin 2相互作用的候选蛋白,交叉后通过分子对接和CO-IP实验验证相互作用关系。采用菌落形成、细胞免疫荧光、细胞划痕和3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四唑(MTT)试验预测操纵候选PPI的临床治疗效果。结果:通过开发三种PPI分类模型并分析超过13,000个数据集,我们确定了3733个以前未知的与自噬相关的PPI。我们的网络分析揭示了Beclin 2在自噬调节中的核心作用,揭示了它与39种新发现的蛋白质的相互作用。值得注意的是,CO-IP研究确定了Beclin 2和Ubiquilin 1之间的实质性相互作用,这是我们的模型所预测的,并在免疫沉淀-质谱分析结果中发现。随后,体外研究表明,在MDA-MB-231细胞中,过表达Beclin 2可增加泛素1,促进自噬依赖性细胞死亡,抑制增殖和转移。结论:本研究不仅提高了我们对TNBC中自噬调控的理解,而且确定了Beclin 2-Ubiquilin 1轴作为精准治疗的一个有希望的靶点。这些发现为药物发现开辟了新的途径,并为这种侵袭性癌症亚型的更有效治疗提供了灵感。
{"title":"Deep learning identification of novel autophagic protein-protein interactions and experimental validation of Beclin 2-Ubiquilin 1 axis in triple-negative breast cancer.","authors":"Xiang Li, Wenke Jin, Lifeng Wu, Huan Wang, Xin Xie, Wei Huang, B O Liu","doi":"10.32604/or.2024.055921","DOIUrl":"10.32604/or.2024.055921","url":null,"abstract":"<p><strong>Background: </strong>Triple-negative breast cancer (TNBC), characterized by its lack of traditional hormone receptors and HER2, presents a significant challenge in oncology due to its poor response to conventional therapies. Autophagy is an important process for maintaining cellular homeostasis, and there are currently autophagy biomarkers that play an effective role in the clinical treatment of tumors. In contrast to targeting protein activity, intervention with protein-protein interaction (PPI) can avoid unrelated crosstalk and regulate the autophagy process with minimal interference pathways.</p><p><strong>Methods: </strong>Here, we employed Naive Bayes, Decision Tree, and k-Nearest Neighbors to elucidate the complex PPI network associated with autophagy in TNBC, aiming to uncover novel therapeutic targets. Meanwhile, the candidate proteins interacting with Beclin 2 were initially screened in MDA-MB-231 cells using Beclin 2 as bait protein by immunoprecipitation-mass spectrometry assay, and the interaction relationship was verified by molecular docking and CO-IP experiments after intersection. Colony formation, cellular immunofluorescence, cell scratch and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) tests were used to predict the clinical therapeutic effects of manipulating candidate PPI.</p><p><strong>Results: </strong>By developing three PPI classification models and analyzing over 13,000 datasets, we identified 3733 previously unknown autophagy-related PPIs. Our network analysis revealed the central role of Beclin 2 in autophagy regulation, uncovering its interactions with 39 newly identified proteins. Notably, the CO-IP studies identified the substantial interaction between Beclin 2 and Ubiquilin 1, which was anticipated by our model and discovered in immunoprecipitation-mass spectrometry assay results. Subsequently, <i>in vitro</i> investigations showed that overexpressing Beclin 2 increased Ubiquilin 1, promoted autophagy-dependent cell death, and inhibited proliferation and metastasis in MDA-MB-231 cells.</p><p><strong>Conclusions: </strong>This study not only enhances our understanding of autophagy regulation in TNBC but also identifies the Beclin 2-Ubiquilin 1 axis as a promising target for precision therapy. These findings open new avenues for drug discovery and offer inspiration for more effective treatments for this aggressive cancer subtype.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"67-81"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671618/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903392","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
SBL-JP-0004: A promising dual inhibitor of JAK2 and PI3KCD against gastric cancer. SBL-JP-0004:一种有前景的JAK2和PI3KCD双重抑制剂,用于胃癌治疗。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.055677
Hassan M Otifi

Background: Gastric cancer (GC) remains a global health burden and is often characterized by heterogeneous molecular profiles and resistance to conventional therapies. The phosphoinositide 3-kinase and PI3K and Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathways play pivotal roles in GC progression, making them attractive targets for therapeutic interventions.

Methods: This study applied a computational and molecular dynamics simulation approach to identify and characterize SBL-JP-0004 as a potential dual inhibitor of JAK2 and PI3KCD kinases. KATOIII and SNU-5 GC cells were used for in vitro evaluation.

Results: SBL-JP-0004 exhibited a robust binding affinity for JAK2 and PI3KCD kinases, as evidenced by molecular docking scores and molecular dynamics simulations. Binding interactions and Gibbs binding free energy estimates confirmed stable and favorable interactions with target proteins. SBL-JP-0004 displayed an half-maximal inhibitory concentration (IC50) value of 118.9 nM against JAK2 kinase and 200.9 nM against PI3KCD enzymes. SBL-JP-0004 exhibited potent inhibition of cell proliferation in KATOIII and SNU-5 cells, with half-maximal growth inhibitory concentration (GI50) values of 250.8 and 516.3 nM, respectively. A significant elevation in the early phase apoptosis (28.53% in KATOIII cells and 26.85% in SNU-5 cells) and late phase apoptosis (17.37% in KATOIII cells and 10.05% in SNU-5 cells) were observed with SBL-JP-0004 treatment compared to 2.1% and 2.83% in their respective controls.

Conclusion: The results highlight SBL-JP-0004 as a promising dual inhibitor targeting JAK2 and PI3KCD kinases for treating GC and warrant further preclinical and clinical investigations to validate its utility in clinical settings.

背景:胃癌(GC)仍然是一个全球性的健康负担,通常以异质性分子谱和对常规治疗的耐药性为特征。磷酸肌苷3-激酶、PI3K和Janus激酶(JAK)信号转导和转录激活因子(JAK- stat)通路在GC进展中起关键作用,使它们成为治疗干预的有吸引力的靶点。方法:本研究采用计算和分子动力学模拟方法鉴定和表征SBL-JP-0004作为JAK2和PI3KCD激酶的潜在双重抑制剂。体外评价采用KATOIII和SNU-5 GC细胞。结果:SBL-JP-0004对JAK2和PI3KCD激酶表现出强大的结合亲和力,分子对接评分和分子动力学模拟证明了这一点。结合相互作用和吉布斯结合自由能估计证实了与靶蛋白的稳定和有利的相互作用。SBL-JP-0004对JAK2激酶和PI3KCD酶的半数最大抑制浓度(IC50)分别为118.9 nM和200.9 nM。SBL-JP-0004对KATOIII和SNU-5细胞的增殖有较强的抑制作用,半最大生长抑制浓度(GI50)分别为250.8 nM和516.3 nM。ssl - jp -0004显著提高了早期凋亡(KATOIII细胞28.53%,SNU-5细胞26.85%)和晚期凋亡(KATOIII细胞17.37%,SNU-5细胞10.05%),而对照组分别为2.1%和2.83%。结论:SBL-JP-0004是一种有前景的靶向JAK2和PI3KCD激酶治疗GC的双重抑制剂,需要进一步的临床前和临床研究来验证其在临床环境中的效用。
{"title":"SBL-JP-0004: A promising dual inhibitor of JAK2 and PI3KCD against gastric cancer.","authors":"Hassan M Otifi","doi":"10.32604/or.2024.055677","DOIUrl":"10.32604/or.2024.055677","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) remains a global health burden and is often characterized by heterogeneous molecular profiles and resistance to conventional therapies. The phosphoinositide 3-kinase and PI3K and Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathways play pivotal roles in GC progression, making them attractive targets for therapeutic interventions.</p><p><strong>Methods: </strong>This study applied a computational and molecular dynamics simulation approach to identify and characterize SBL-JP-0004 as a potential dual inhibitor of JAK2 and PI3KCD kinases. KATOIII and SNU-5 GC cells were used for <i>in vitro</i> evaluation.</p><p><strong>Results: </strong>SBL-JP-0004 exhibited a robust binding affinity for JAK2 and PI3KCD kinases, as evidenced by molecular docking scores and molecular dynamics simulations. Binding interactions and Gibbs binding free energy estimates confirmed stable and favorable interactions with target proteins. SBL-JP-0004 displayed an half-maximal inhibitory concentration (IC<sub>50</sub>) value of 118.9 nM against JAK2 kinase and 200.9 nM against PI3KCD enzymes. SBL-JP-0004 exhibited potent inhibition of cell proliferation in KATOIII and SNU-5 cells, with half-maximal growth inhibitory concentration (GI<sub>50</sub>) values of 250.8 and 516.3 nM, respectively. A significant elevation in the early phase apoptosis (28.53% in KATOIII cells and 26.85% in SNU-5 cells) and late phase apoptosis (17.37% in KATOIII cells and 10.05% in SNU-5 cells) were observed with SBL-JP-0004 treatment compared to 2.1% and 2.83% in their respective controls.</p><p><strong>Conclusion: </strong>The results highlight SBL-JP-0004 as a promising dual inhibitor targeting JAK2 and PI3KCD kinases for treating GC and warrant further preclinical and clinical investigations to validate its utility in clinical settings.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"235-243"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671411/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903042","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
CES1 is associated with cisplatin resistance and poor prognosis of head and neck squamous cell carcinoma. CES1 与头颈部鳞状细胞癌的顺铂耐药性和不良预后有关。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.052244
Chuan Jiang, Chunlei Liu, X I Yao, Jingya Su, Wei Lu, Zhengbo Wei, Ying Xie

Background: Head and neck squamous cell carcinoma (HNSCC) is a prevalent form of cancer globally, with chemoresistance posing a major challenge in treatment outcomes. The efficacy of the commonly used chemotherapeutic agent, cisplatin, is diminished in patients with poor prognoses.

Methods: Various bioinformatics databases were utilized to examine Carboxylesterase 1 (CES1) gene expression, clinicopathologic features, patient survival analysis, and gene function. An organoid model of HNSCC was established, along with the induction of drug-resistant HNSCC in the organoid model. CES1 expression was assessed using qRT-PCR and Western Blot, and differential markers were identified through transcriptome sequencing. Knockdown and overexpression models of CES1 were created in SCC-9 and patient-derived organoid (PDO) cells using shRNA and lentivirus to investigate the tumor biology and cisplatin resistance associated with CES1.

Results: Research in bioinformatics has uncovered a strong correlation between the expression level of CES1 and the prognosis of HNSCC. The data suggests a significant link between CES1 expression and tobacco smoking. RNA-sequencing revealed a notable increase in CES1 expression in HNSCC-PDOcis-R cells compared to the parental PDO cells. Subsequently, we performed in vitro studies by HNSCC-PDO and SCC-9 and found that CES1-overexpressing cells exhibited reduced sensitivity to cisplatin and stronger tumor malignant biological behavior compared with CES1-knockdown cells.

Conclusion: The observed association between CES1 expression and tobacco smoking implies a potential influence of smoking on the efficacy of cisplatin-based chemotherapy in HNSCC through the regulation of CES1 expression.

背景:头颈部鳞状细胞癌(HNSCC)是一种全球流行的癌症,其化疗耐药性是治疗效果的一大挑战。常用化疗药物顺铂在预后不良的患者中疗效减弱:方法:利用各种生物信息学数据库研究羧基酯酶1(CES1)基因表达、临床病理特征、患者生存分析和基因功能。建立了HNSCC的类器官模型,并在类器官模型中诱导出耐药的HNSCC。利用 qRT-PCR 和 Western Blot 评估了 CES1 的表达,并通过转录组测序确定了差异标记。利用 shRNA 和慢病毒在 SCC-9 和患者衍生类器官(PDO)细胞中创建了 CES1 的敲除和过表达模型,以研究与 CES1 相关的肿瘤生物学和顺铂耐药性:生物信息学研究发现,CES1 的表达水平与 HNSCC 的预后密切相关。数据表明,CES1的表达与吸烟有重要关系。RNA测序显示,与亲本PDO细胞相比,HNSCC-PDOcis-R细胞中CES1的表达明显增加。随后,我们用 HNSCC-PDO 和 SCC-9 进行了体外研究,发现与 CES1 敲除的细胞相比,CES1 表达的细胞对顺铂的敏感性降低,肿瘤恶性生物学行为更强:结论:观察到的CES1表达与吸烟之间的关联意味着,吸烟可能会通过调控CES1的表达影响HNSCC中以顺铂为基础的化疗的疗效。
{"title":"CES1 is associated with cisplatin resistance and poor prognosis of head and neck squamous cell carcinoma.","authors":"Chuan Jiang, Chunlei Liu, X I Yao, Jingya Su, Wei Lu, Zhengbo Wei, Ying Xie","doi":"10.32604/or.2024.052244","DOIUrl":"10.32604/or.2024.052244","url":null,"abstract":"<p><strong>Background: </strong>Head and neck squamous cell carcinoma (HNSCC) is a prevalent form of cancer globally, with chemoresistance posing a major challenge in treatment outcomes. The efficacy of the commonly used chemotherapeutic agent, cisplatin, is diminished in patients with poor prognoses.</p><p><strong>Methods: </strong>Various bioinformatics databases were utilized to examine Carboxylesterase 1 (CES1) gene expression, clinicopathologic features, patient survival analysis, and gene function. An organoid model of HNSCC was established, along with the induction of drug-resistant HNSCC in the organoid model. CES1 expression was assessed using qRT-PCR and Western Blot, and differential markers were identified through transcriptome sequencing. Knockdown and overexpression models of CES1 were created in SCC-9 and patient-derived organoid (PDO) cells using shRNA and lentivirus to investigate the tumor biology and cisplatin resistance associated with CES1.</p><p><strong>Results: </strong>Research in bioinformatics has uncovered a strong correlation between the expression level of CES1 and the prognosis of HNSCC. The data suggests a significant link between CES1 expression and tobacco smoking. RNA-sequencing revealed a notable increase in CES1 expression in HNSCC-PDO<sup>cis-R</sup> cells compared to the parental PDO cells. Subsequently, we performed <i>in vitro</i> studies by HNSCC-PDO and SCC-9 and found that CES1-overexpressing cells exhibited reduced sensitivity to cisplatin and stronger tumor malignant biological behavior compared with CES1-knockdown cells.</p><p><strong>Conclusion: </strong>The observed association between CES1 expression and tobacco smoking implies a potential influence of smoking on the efficacy of cisplatin-based chemotherapy in HNSCC through the regulation of CES1 expression.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1935-1948"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576922/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LncRNA AFAP1-AS1 exhibits oncogenic characteristics and promotes gemcitabine-resistance of cervical cancer cells through miR-7-5p/EGFR axis. LncRNA AFAP1-AS1 具有致癌特性,并通过 miR-7-5p/EGFR 轴促进宫颈癌细胞对吉西他滨的耐药性。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.044547
Chaoqun Wang, Ting Zhang, Chaohe Zhang

Background: Drug resistance is the main factor contributing to cancer recurrence and poor prognosis. Exploration of drug resistance-related mechanisms and effective therapeutic targets are the aim of molecular targeted therapy. In our study, the role of long non-coding RNA (lncRNA) AFAP1-AS1 in gemcitabine resistance and related mechanisms were explored in cervical cancer cells.

Methods: Gemcitabine-resistant cervical cancer cell lines HT-3-Gem and SW756-Gem were constructed using the gemcitabine concentration gradient method. The overall survival rates and recurrence-free survival rates were evaluated by Kaplan-Meier analysis. The interaction was verified through a Dual-luciferase reporter gene assay and a Biotinylated RNA pull-down assay. Cell proliferation ability was assessed through methyl-thiazolyl-tetrazolium (MTT), soft agar, and colony formation experiments. Cell cycle and apoptosis were detected by flow cytometry.

Results: Up-regulation of AFAP1-AS1 in cervical cancer predicted a poor prognosis. Besides, patients in the gemcitabine-resistance group had higher levels of AFAP1-AS1 than the gemcitabine-sensitive group. AFAP1-AS1 promoted tumor growth and induced gemcitabine tolerance of cervical cancer cells. In addition, AFAP1-AS1 mediated epidermal growth factor receptor (EGFR) expression by serving as a molecular sponge for microRNA-7a-5p (miR-7-5p). This present study also proved that the knockdown of EGFR or overexpression of miR-7a-5p abolished the accelerative role of AFAP1-AS1 overexpression in cancer progression and gemcitabine tolerance.

Conclusions: In general, the AFAP1-AS1/miR-7-5p/EGFR axis was tightly related to the progression and gemcitabine tolerance of cervical cancer, providing potential targets for the management of cervical cancer.

背景:耐药性是导致癌症复发和预后不良的主要因素。探索耐药相关机制和有效治疗靶点是分子靶向治疗的目标。本研究探讨了宫颈癌细胞中长非编码 RNA(lncRNA)AFAP1-AS1 在吉西他滨耐药中的作用及相关机制:方法:采用吉西他滨浓度梯度法构建了吉西他滨耐药的宫颈癌细胞系HT-3-Gem和SW756-Gem。采用 Kaplan-Meier 分析法评估了总生存率和无复发生存率。通过双荧光素酶报告基因试验和生物素化 RNA 拉取试验验证了相互作用。细胞增殖能力通过甲基-噻唑基-四唑啉(MTT)、软琼脂和菌落形成实验进行评估。流式细胞术检测细胞周期和细胞凋亡:结果:AFAP1-AS1在宫颈癌中的上调预示着预后不良。此外,吉西他滨耐药组患者的AFAP1-AS1水平高于吉西他滨敏感组。AFAP1-AS1能促进肿瘤生长,诱导宫颈癌细胞对吉西他滨耐受。此外,AFAP1-AS1通过充当microRNA-7a-5p(miR-7-5p)的分子海绵,介导表皮生长因子受体(EGFR)的表达。本研究还证明,敲除表皮生长因子受体或过表达 miR-7a-5p 可消除 AFAP1-AS1 过表达在癌症进展和吉西他滨耐受中的加速作用:总的来说,AFAP1-AS1/miR-7-5p/EGFR轴与宫颈癌的进展和吉西他滨耐受性密切相关,为宫颈癌的治疗提供了潜在靶点。
{"title":"LncRNA AFAP1-AS1 exhibits oncogenic characteristics and promotes gemcitabine-resistance of cervical cancer cells through miR-7-5p/EGFR axis.","authors":"Chaoqun Wang, Ting Zhang, Chaohe Zhang","doi":"10.32604/or.2024.044547","DOIUrl":"10.32604/or.2024.044547","url":null,"abstract":"<p><strong>Background: </strong>Drug resistance is the main factor contributing to cancer recurrence and poor prognosis. Exploration of drug resistance-related mechanisms and effective therapeutic targets are the aim of molecular targeted therapy. In our study, the role of long non-coding RNA (lncRNA) AFAP1-AS1 in gemcitabine resistance and related mechanisms were explored in cervical cancer cells.</p><p><strong>Methods: </strong>Gemcitabine-resistant cervical cancer cell lines HT-3-Gem and SW756-Gem were constructed using the gemcitabine concentration gradient method. The overall survival rates and recurrence-free survival rates were evaluated by Kaplan-Meier analysis. The interaction was verified through a Dual-luciferase reporter gene assay and a Biotinylated RNA pull-down assay. Cell proliferation ability was assessed through methyl-thiazolyl-tetrazolium (MTT), soft agar, and colony formation experiments. Cell cycle and apoptosis were detected by flow cytometry.</p><p><strong>Results: </strong>Up-regulation of AFAP1-AS1 in cervical cancer predicted a poor prognosis. Besides, patients in the gemcitabine-resistance group had higher levels of AFAP1-AS1 than the gemcitabine-sensitive group. AFAP1-AS1 promoted tumor growth and induced gemcitabine tolerance of cervical cancer cells. In addition, AFAP1-AS1 mediated epidermal growth factor receptor (EGFR) expression by serving as a molecular sponge for microRNA-7a-5p (miR-7-5p). This present study also proved that the knockdown of EGFR or overexpression of miR-7a-5p abolished the accelerative role of AFAP1-AS1 overexpression in cancer progression and gemcitabine tolerance.</p><p><strong>Conclusions: </strong>In general, the AFAP1-AS1/miR-7-5p/EGFR axis was tightly related to the progression and gemcitabine tolerance of cervical cancer, providing potential targets for the management of cervical cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1867-1879"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576921/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687903","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 superiority of PMFs on reversing drug resistance of colon cancer and the effect on aerobic glycolysis-ROS-autophagy signaling axis. PMFs 在逆转结肠癌耐药性方面的优势以及对有氧糖酵解-ROS-自噬信号轴的影响。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.048778
Yuqin Yin, Y U Wu, Hongliang Huang, Yingying Duan, Zhongwen Yuan, Lihui Cao, Jinjin Ying, Yongheng Zhou, Senling Feng

Background: Polymethoxylated flavones (PMFs) are compounds present in citrus peels and other Rutaceae plants, which exhibit diverse biological activities, including robust antitumor and antioxidant effects. However, the mechanism of PMFs in reversing drug resistance to colon cancer remains unknown. In the present study, we aimed to investigate the potential connection between the aerobic glycolysis-ROS-autophagy signaling axis and the reversal of PTX resistance in colon cancer by PMFs.

Methods: MTT Cell viability assay and colony formation assay were used to investigate the effect of PMFs combined with PTX in reversing HCT8/T cell resistance ex vivo; the mRNA and protein levels of the target were detected by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis), quantitative real-time fluorescence polymerase chain reaction (qRT-PCR) and Western blot protein immunoblotting (WB); An HCT8/T cell xenograft model was established to investigate the MDR reversal activity of PMFs in vivo; The extracellular acidification rate (ECAR) and the oxygen consumption rate (OCR) were detected to assess the cellular oxygen consumption rate and glycolytic process.

Results: HCT8/T cells demonstrated significant resistance to PTX, up-regulating the expression levels of ABCB1 mRNA, P-gp, LC3-I, and LC3-II protein, and increasing intracellular reactive oxygen species (ROS) content. PMFs mainly contain two active ingredients, nobiletin, and tangeretin, which were able to reverse drug resistance in HCT8/T cells in a concentration-dependent manner. PMFs exhibited high tolerance in the HCT8/T nude mouse model while increasing the sensitivity of PTX-resistant cells and suppressing tumor growth significantly. PMFs combined with PTX reduced extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) in HCT8/T cells. Additionally, PMFs reduced intracellular ROS content, down-regulated the expression levels of autophagy-related proteins LC3-I, LC3-II, Beclin1, and ATG7, and significantly reduced the number of autophagosomes in HCT8/T cells.

Conclusions: The present study demonstrated that PMFs could potentially reverse PTX resistance in colon cancer by regulating the aerobic glycolysis-ROS-autophagy signaling axis, which indicated that PMFs would be potential potentiators for future chemotherapeutic agents in colon cancer.

背景:聚甲氧基黄酮(PMFs)是存在于柑橘皮和其他芸香科植物中的化合物,具有多种生物活性,包括强大的抗肿瘤和抗氧化作用。然而,PMFs 在逆转结肠癌耐药性方面的作用机制尚不清楚。在本研究中,我们旨在探讨有氧糖酵解-ROS-自噬信号轴与PMFs逆转结肠癌PTX耐药性之间的潜在联系:方法:采用MTT细胞活力检测和菌落形成检测研究PMFs联合PTX对HCT8/T细胞体内耐药性的逆转作用;采用SDS-PAGE(十二烷基硫酸钠-聚丙烯酰胺凝胶电泳)、实时荧光定量聚合酶链式反应(qRT-PCR)和Western blot蛋白免疫印迹(WB)检测靶标的mRNA和蛋白水平;建立HCT8/T细胞异种移植模型,研究PMFs在体内的MDR逆转活性;检测细胞外酸化率(ECAR)和耗氧量(OCR),评估细胞耗氧量和糖酵解过程。结果HCT8/T细胞对PTX表现出明显的耐药性,上调了ABCB1 mRNA、P-gp、LC3-I和LC3-II蛋白的表达水平,增加了细胞内活性氧(ROS)含量。PMFs主要含有两种活性成分:金霉素和桔皮苷,它们能够以浓度依赖性的方式逆转HCT8/T细胞的耐药性。PMFs在HCT8/T裸鼠模型中表现出较高的耐受性,同时提高了PTX耐药细胞的敏感性,并显著抑制了肿瘤的生长。PMFs 与 PTX 联用可降低 HCT8/T 细胞的细胞外酸化率(ECAR)和耗氧量(OCR)。此外,PMFs还能降低细胞内ROS含量,下调自噬相关蛋白LC3-I、LC3-II、Beclin1和ATG7的表达水平,并显著减少HCT8/T细胞中自噬体的数量:本研究表明,PMFs可通过调节有氧糖酵解-ROS-自噬信号轴逆转结肠癌对PTX的耐药性,这表明PMFs将成为未来结肠癌化疗药物的潜在增效剂。
{"title":"The superiority of PMFs on reversing drug resistance of colon cancer and the effect on aerobic glycolysis-ROS-autophagy signaling axis.","authors":"Yuqin Yin, Y U Wu, Hongliang Huang, Yingying Duan, Zhongwen Yuan, Lihui Cao, Jinjin Ying, Yongheng Zhou, Senling Feng","doi":"10.32604/or.2024.048778","DOIUrl":"10.32604/or.2024.048778","url":null,"abstract":"<p><strong>Background: </strong>Polymethoxylated flavones (PMFs) are compounds present in citrus peels and other Rutaceae plants, which exhibit diverse biological activities, including robust antitumor and antioxidant effects. However, the mechanism of PMFs in reversing drug resistance to colon cancer remains unknown. In the present study, we aimed to investigate the potential connection between the aerobic glycolysis-ROS-autophagy signaling axis and the reversal of PTX resistance in colon cancer by PMFs.</p><p><strong>Methods: </strong>MTT Cell viability assay and colony formation assay were used to investigate the effect of PMFs combined with PTX in reversing HCT8/T cell resistance <i>ex vivo</i>; the mRNA and protein levels of the target were detected by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis), quantitative real-time fluorescence polymerase chain reaction (qRT-PCR) and Western blot protein immunoblotting (WB); An HCT8/T cell xenograft model was established to investigate the MDR reversal activity of PMFs <i>in vivo</i>; The extracellular acidification rate (ECAR) and the oxygen consumption rate (OCR) were detected to assess the cellular oxygen consumption rate and glycolytic process.</p><p><strong>Results: </strong>HCT8/T cells demonstrated significant resistance to PTX, up-regulating the expression levels of ABCB1 mRNA, P-gp, LC3-I, and LC3-II protein, and increasing intracellular reactive oxygen species (ROS) content. PMFs mainly contain two active ingredients, nobiletin, and tangeretin, which were able to reverse drug resistance in HCT8/T cells in a concentration-dependent manner. PMFs exhibited high tolerance in the HCT8/T nude mouse model while increasing the sensitivity of PTX-resistant cells and suppressing tumor growth significantly. PMFs combined with PTX reduced extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) in HCT8/T cells. Additionally, PMFs reduced intracellular ROS content, down-regulated the expression levels of autophagy-related proteins LC3-I, LC3-II, Beclin1, and ATG7, and significantly reduced the number of autophagosomes in HCT8/T cells.</p><p><strong>Conclusions: </strong>The present study demonstrated that PMFs could potentially reverse PTX resistance in colon cancer by regulating the aerobic glycolysis-ROS-autophagy signaling axis, which indicated that PMFs would be potential potentiators for future chemotherapeutic agents in colon cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1891-1902"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687908","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
Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature. 利用多指标分析探索胶质瘤诊断工具并优化基于交叉基因特征的药物治疗选择
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.046191
Yushi Yang, Chujiao Hu, Shan Lei, Xin Bao, Zhirui Zeng, Wenpeng Cao

Background: The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma.

Materials and methods: The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments.

Results: A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (POSTN), chitinase 3 like 1 (CHI3L1), serum amyloid A1 (SAA1), and matrix metallopeptidase 9 (MMP9) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib.

Conclusions: In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.

背景:胶质瘤患者的预后和治疗获益的异质性是由肿瘤微环境特征造成的。然而,能反映胶质瘤微环境特征并预测其预后的生物标志物非常有限。因此,我们旨在开发一种能有效预测预后、区分微环境特征并优化胶质瘤患者药物选择的模型:采用CIBERSORT算法、批量测序分析和单细胞RNA(scRNA)分析来确定胶质瘤组织中M2巨噬细胞和癌细胞之间的重要交叉基因。根据交叉对话基因的表达构建了预测模型,并在多个队列中验证了该模型对预后、复发预测和微环境特征的影响。利用OncoPredict算法和相关的细胞生物学实验评估了预测模型对药物选择的影响:结果:胶质瘤组织中M2巨噬细胞的高丰度预示着预后不良,巨噬细胞和癌细胞之间的交叉对话在肿瘤微环境的形成中起着至关重要的作用。研究发现了 8 个参与巨噬细胞和癌细胞之间交叉对话的基因。在这些基因中,我们选择了骨膜增生蛋白(POSTN)、几丁质酶 3 like 1(CHI3L1)、血清淀粉样蛋白 A1(SAA1)和基质金属肽酶 9(MMP9)来构建预测模型。所建立的模型在区分患者预后、复发病例以及高炎症、缺氧和免疫抑制等特征方面具有显著疗效。此外,该模型还可作为指导使用曲美替尼的重要工具:综上所述,本研究全面了解了胶质瘤中M2巨噬细胞与癌细胞之间的相互作用;利用交叉对话基因特征建立了一个预测模型,可预测患者预后、复发情况和微环境特征的分化;有助于优化曲美替尼在胶质瘤患者中的应用。
{"title":"Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature.","authors":"Yushi Yang, Chujiao Hu, Shan Lei, Xin Bao, Zhirui Zeng, Wenpeng Cao","doi":"10.32604/or.2024.046191","DOIUrl":"10.32604/or.2024.046191","url":null,"abstract":"<p><strong>Background: </strong>The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma.</p><p><strong>Materials and methods: </strong>The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments.</p><p><strong>Results: </strong>A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (<i>POSTN</i>), chitinase 3 like 1 (<i>CHI3L1</i>), serum amyloid A1 (<i>SAA1</i>), and matrix metallopeptidase 9 (<i>MMP9</i>) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib.</p><p><strong>Conclusions: </strong>In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1921-1934"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687913","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncology Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1