Pub Date : 2024-09-01Epub Date: 2024-08-02DOI: 10.3892/ijo.2024.5676
Shenghao Li, Liyuan Hao, Na Li, Xiaoyu Hu, Huimin Yan, Erhei Dai, Xinli Shi
The Hippo signaling pathway plays a pivotal role in regulating cell growth and organ size. Its regulatory effects on hepatocellular carcinoma (HCC) encompass diverse aspects, including cell proliferation, invasion and metastasis, tumor drug resistance, metabolic reprogramming, immunomodulatory effects and autophagy. Yes‑associated protein 1 (YAP1), a potent transcriptional coactivator and a major downstream target tightly controlled by the Hippo pathway, is influenced by various molecules and pathways. The expression of YAP1 in different cell types within the liver tumor microenvironment exerts varying effects on tumor outcomes, warranting careful consideration. Therefore, research on YAP1‑targeted therapies merits attention. This review discusses the composition and regulation mechanism of the Hippo/YAP1 signaling pathway and its relationship with HCC, offering insights for future research and cancer prevention strategies.
希波信号通路在调节细胞生长和器官大小方面发挥着关键作用。它对肝细胞癌(HCC)的调控作用涉及多个方面,包括细胞增殖、侵袭和转移、肿瘤耐药性、代谢重编程、免疫调节作用和自噬。YAP1(Yes-associated protein 1,YAP1)是一种有效的转录辅激活因子,也是受Hippo通路严格控制的主要下游靶标,它受多种分子和通路的影响。YAP1在肝脏肿瘤微环境中不同细胞类型中的表达对肿瘤结果的影响各不相同,值得仔细研究。因此,有关 YAP1 靶向疗法的研究值得关注。本综述讨论了 Hippo/YAP1 信号通路的组成和调控机制及其与 HCC 的关系,为未来的研究和癌症预防策略提供启示。
{"title":"Targeting the Hippo/YAP1 signaling pathway in hepatocellular carcinoma: From mechanisms to therapeutic drugs (Review).","authors":"Shenghao Li, Liyuan Hao, Na Li, Xiaoyu Hu, Huimin Yan, Erhei Dai, Xinli Shi","doi":"10.3892/ijo.2024.5676","DOIUrl":"10.3892/ijo.2024.5676","url":null,"abstract":"<p><p>The Hippo signaling pathway plays a pivotal role in regulating cell growth and organ size. Its regulatory effects on hepatocellular carcinoma (HCC) encompass diverse aspects, including cell proliferation, invasion and metastasis, tumor drug resistance, metabolic reprogramming, immunomodulatory effects and autophagy. Yes‑associated protein 1 (YAP1), a potent transcriptional coactivator and a major downstream target tightly controlled by the Hippo pathway, is influenced by various molecules and pathways. The expression of YAP1 in different cell types within the liver tumor microenvironment exerts varying effects on tumor outcomes, warranting careful consideration. Therefore, research on YAP1‑targeted therapies merits attention. This review discusses the composition and regulation mechanism of the Hippo/YAP1 signaling pathway and its relationship with HCC, offering insights for future research and cancer prevention strategies.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 3","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302957/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141874778","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Subsequently to the publication of the above article, an interested reader drew to the authors' attention that certain of the EdU assay data shown in Fig. 7E on p. 2418 had already appeared in different form in a previously published paper written by different authors at different research institutes. Owing to the fact that the contentious data in the above article had already been published prior to its submission to International Journal of Oncology, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [International Journal of Oncology 53: 2409‑2422, 2018; DOI: 10.3892/ijo.2018.4586].
在上述文章发表后,一位感兴趣的读者提请作者注意,第 2418 页图 7E 中显示的某些 EdU 检测数据已经以不同的形式出现在以前发表的一篇论文中,该论文由不同的作者在不同的研究机构撰写。由于上述文章中有争议的数据在提交给《国际肿瘤学杂志》之前已经发表,因此编辑决定从该杂志上撤下这篇论文。已要求作者就这些问题做出解释,但编辑部没有收到回复。对于给读者带来的不便,编辑深表歉意。 [International Journal of Oncology 53: 2409-2422, 2018; DOI: 10.3892/ijo.2018.4586]。
{"title":"[Retracted] Mst1 regulates non‑small cell lung cancer A549 cell apoptosis by inducing mitochondrial damage via ROCK1/F‑actin pathways.","authors":"Weiqiang Zhang, Keiqiang Liu, Yingxin Pei, Jingbo Ma, Jiang Tan, Jing Zhao","doi":"10.3892/ijo.2024.5663","DOIUrl":"10.3892/ijo.2024.5663","url":null,"abstract":"<p><p>Subsequently to the publication of the above article, an interested reader drew to the authors' attention that certain of the EdU assay data shown in Fig. 7E on p. 2418 had already appeared in different form in a previously published paper written by different authors at different research institutes. Owing to the fact that the contentious data in the above article had already been published prior to its submission to <i>International Journal of Oncology</i>, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [International Journal of Oncology 53: 2409‑2422, 2018; DOI: 10.3892/ijo.2018.4586].</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141317268","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-01Epub Date: 2024-07-12DOI: 10.3892/ijo.2024.5668
Jonathan H Sherman, Adam Bobak, Tasneem Arsiwala, Paul Lockman, Sonikpreet Aulakh
Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. The current standard of care includes surgery, radiation therapy, temozolomide; and tumor‑treating fields leads to dismal overall survival. There are far limited treatments upon recurrence. Therapies to date are ineffective as a result of several factors, including the presence of the blood‑brain barrier, blood tumor barrier, glioma stem‑like cells and genetic heterogeneity in GBM. In the present review, the potential mechanisms that lead to treatment resistance in GBM and the measures which have been taken so far to attempt to overcome the resistance were discussed. The complex biology of GBM and lack of comprehensive understanding of the development of therapeutic resistance in GBM demands discovery of novel antigens that are targetable and provide effective therapeutic strategies.
{"title":"Targeting drug resistance in glioblastoma (Review).","authors":"Jonathan H Sherman, Adam Bobak, Tasneem Arsiwala, Paul Lockman, Sonikpreet Aulakh","doi":"10.3892/ijo.2024.5668","DOIUrl":"10.3892/ijo.2024.5668","url":null,"abstract":"<p><p>Glioblastoma (GBM) is the most common malignancy of the central nervous system in adults. The current standard of care includes surgery, radiation therapy, temozolomide; and tumor‑treating fields leads to dismal overall survival. There are far limited treatments upon recurrence. Therapies to date are ineffective as a result of several factors, including the presence of the blood‑brain barrier, blood tumor barrier, glioma stem‑like cells and genetic heterogeneity in GBM. In the present review, the potential mechanisms that lead to treatment resistance in GBM and the measures which have been taken so far to attempt to overcome the resistance were discussed. The complex biology of GBM and lack of comprehensive understanding of the development of therapeutic resistance in GBM demands discovery of novel antigens that are targetable and provide effective therapeutic strategies.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251740/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141590310","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-01Epub Date: 2024-06-14DOI: 10.3892/ijo.2024.5664
Shouying Xu, Chongying Zhu, Qiang Xu, Zihao An, Shu Xu, Ge Xuan, Chao Lin, Chao Tang
Genes encoding subunits of SWI/SNF (BAF) chromatin‑remodeling complexes are recurrently mutated in a broad array of tumor types, and among the subunits, ARID1A is the most frequent target with mutations. In the present study, it was reported that ARID1A inhibits the epithelial‑mesenchymal transition (EMT) and stemness of ovarian cancer cells, accompanied by reduced cell viability, migration and colony formation, suggesting that ARID1A acts as a tumor suppressor in ovarian cancer. Mechanistically, ARID1A exerts its inhibitory effects on ovarian cancer cells by activating the Hippo signaling pathway. Conversely, the overexpression of a gain‑of‑function transcriptional co‑activator with PDZ‑binding motif (TAZ) mutant (TAZ‑Ser89) effectively reverses the effects induced by ARID1A. In addition, activation of Hippo signaling apparently upregulates ARID1A protein expression, whereas ectopic expression of TAZ‑Ser89 results in the markedly decreased ARID1A levels, indicating a feedback of ARID1A‑TAZ in regulating ovarian cancer cell EMT and stemness. Thus, the present study uncovered the role of ARID1A through the Hippo/TAZ pathway in modulating EMT and stemness of ovarian cancer cells, and providing with evidence that TAZ inhibitors could effectively prevent initiation and metastasis of ovarian cancer cases where ARID1A is lost or mutated.
{"title":"ARID1A restrains EMT and stemness of ovarian cancer cells through the Hippo pathway.","authors":"Shouying Xu, Chongying Zhu, Qiang Xu, Zihao An, Shu Xu, Ge Xuan, Chao Lin, Chao Tang","doi":"10.3892/ijo.2024.5664","DOIUrl":"10.3892/ijo.2024.5664","url":null,"abstract":"<p><p>Genes encoding subunits of SWI/SNF (BAF) chromatin‑remodeling complexes are recurrently mutated in a broad array of tumor types, and among the subunits, ARID1A is the most frequent target with mutations. In the present study, it was reported that ARID1A inhibits the epithelial‑mesenchymal transition (EMT) and stemness of ovarian cancer cells, accompanied by reduced cell viability, migration and colony formation, suggesting that ARID1A acts as a tumor suppressor in ovarian cancer. Mechanistically, ARID1A exerts its inhibitory effects on ovarian cancer cells by activating the Hippo signaling pathway. Conversely, the overexpression of a gain‑of‑function transcriptional co‑activator with PDZ‑binding motif (TAZ) mutant (TAZ‑Ser89) effectively reverses the effects induced by ARID1A. In addition, activation of Hippo signaling apparently upregulates ARID1A protein expression, whereas ectopic expression of TAZ‑Ser89 results in the markedly decreased ARID1A levels, indicating a feedback of ARID1A‑TAZ in regulating ovarian cancer cell EMT and stemness. Thus, the present study uncovered the role of ARID1A through the Hippo/TAZ pathway in modulating EMT and stemness of ovarian cancer cells, and providing with evidence that TAZ inhibitors could effectively prevent initiation and metastasis of ovarian cancer cases where ARID1A is lost or mutated.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251745/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141317269","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-01Epub Date: 2024-06-14DOI: 10.3892/ijo.2024.5665
Siyuan Wu, Qi Zhao, Shengjuan Liu, Jiayang Kuang, Ji Zhang, Annabeth Onga, Yiwei Shen, Jiaying Wang, Hehuan Sui, Lianli Ni, Yuxin Ye, Xinyue Tu, Han-Bo Le, Yihu Zheng, Ri Cui, Wangyu Zhu
Non‑small cell lung cancer (NSCLC) is one of the major causes of cancer‑related death worldwide. Cisplatin is a front‑line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin‑mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription‑quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS‑mediated endoplasmic reticulum stress, and the C‑Jun‑amino‑terminal kinase and p38 mitogen‑activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS‑mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.
{"title":"Polydatin, a potential NOX5 agonist, synergistically enhances antitumor activity of cisplatin by stimulating oxidative stress in non‑small cell lung cancer.","authors":"Siyuan Wu, Qi Zhao, Shengjuan Liu, Jiayang Kuang, Ji Zhang, Annabeth Onga, Yiwei Shen, Jiaying Wang, Hehuan Sui, Lianli Ni, Yuxin Ye, Xinyue Tu, Han-Bo Le, Yihu Zheng, Ri Cui, Wangyu Zhu","doi":"10.3892/ijo.2024.5665","DOIUrl":"10.3892/ijo.2024.5665","url":null,"abstract":"<p><p>Non‑small cell lung cancer (NSCLC) is one of the major causes of cancer‑related death worldwide. Cisplatin is a front‑line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin‑mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription‑quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS‑mediated endoplasmic reticulum stress, and the C‑Jun‑amino‑terminal kinase and p38 mitogen‑activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS‑mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251743/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141317270","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-01Epub Date: 2024-06-28DOI: 10.3892/ijo.2024.5667
Carlos Jiménez-Cortegana, Cristian Gutiérrez-García, Flora Sánchez-Jiménez, Teresa Vilariño-García, Rocio Flores-Campos, Antonio Pérez-Pérez, Carmen Garnacho, Maria L Sánchez-León, Daniel J García-Domínguez, Lourdes Hontecillas-Prieto, Natalia Palazón-Carrión, Luis De La Cruz-Merino, Víctor Sánchez-Margalet
Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.
{"title":"Impact of obesity‑associated myeloid‑derived suppressor cells on cancer risk and progression (Review).","authors":"Carlos Jiménez-Cortegana, Cristian Gutiérrez-García, Flora Sánchez-Jiménez, Teresa Vilariño-García, Rocio Flores-Campos, Antonio Pérez-Pérez, Carmen Garnacho, Maria L Sánchez-León, Daniel J García-Domínguez, Lourdes Hontecillas-Prieto, Natalia Palazón-Carrión, Luis De La Cruz-Merino, Víctor Sánchez-Margalet","doi":"10.3892/ijo.2024.5667","DOIUrl":"10.3892/ijo.2024.5667","url":null,"abstract":"<p><p>Obesity is a chronic disease caused by the accumulation of excessive adipose tissue. This disorder is characterized by chronic low‑grade inflammation, which promotes the release of proinflammatory mediators, including cytokines, chemokines and leptin. Simultaneously, chronic inflammation can predispose to cancer development, progression and metastasis. Proinflammatory molecules are involved in the recruitment of specific cell populations in the tumor microenvironment. These cell populations include myeloid‑derived suppressor cells (MDSCs), a heterogeneous, immature myeloid population with immunosuppressive abilities. Obesity‑associated MDSCs have been linked with tumor dissemination, progression and poor clinical outcomes. A comprehensive literature review was conducted to assess the impact of obesity‑associated MDSCs on cancer in both preclinical models and oncological patients with obesity. A secondary objective was to examine the key role that leptin, the most important proinflammatory mediator released by adipocytes, plays in MDSC‑driven immunosuppression Finally, an overview is provided of the different therapeutic approaches available to target MDSCs in the context of obesity‑related cancer.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251741/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-01Epub Date: 2024-07-12DOI: 10.3892/ijo.2024.5669
Bangjie Chen, Yipin Yang, Xinyi Wang, Wenzhi Yang, You Lu, Daoyue Wang, Enba Zhuo, Yanchao Tang, Junhong Su, Guozheng Tang, Song Shao, Kangsheng Gu
Cancer is characterized by unlimited proliferation and metastasis, and traditional therapeutic strategies usually result in the acquisition of drug resistance, thus highlighting the need for more personalized treatment. mRNA vaccines transfer the gene sequences of exogenous target antigens into human cells through transcription and translation to stimulate the body to produce specific immune responses against the encoded proteins, so as to enable the body to obtain immune protection against said antigens; this approach may be adopted for personalized cancer therapy. Since the recent coronavirus pandemic, the development of mRNA vaccines has seen substantial progress and widespread adoption. In the present review, the development of mRNA vaccines, their mechanisms of action, factors influencing their function and the current clinical applications of the vaccine are discussed. A focus is placed on the application of mRNA vaccines in cancer, with the aim of highlighting unique advances and the remaining challenges of this novel and promising therapeutic approach.
{"title":"mRNA vaccine development and applications: A special focus on tumors (Review).","authors":"Bangjie Chen, Yipin Yang, Xinyi Wang, Wenzhi Yang, You Lu, Daoyue Wang, Enba Zhuo, Yanchao Tang, Junhong Su, Guozheng Tang, Song Shao, Kangsheng Gu","doi":"10.3892/ijo.2024.5669","DOIUrl":"10.3892/ijo.2024.5669","url":null,"abstract":"<p><p>Cancer is characterized by unlimited proliferation and metastasis, and traditional therapeutic strategies usually result in the acquisition of drug resistance, thus highlighting the need for more personalized treatment. mRNA vaccines transfer the gene sequences of exogenous target antigens into human cells through transcription and translation to stimulate the body to produce specific immune responses against the encoded proteins, so as to enable the body to obtain immune protection against said antigens; this approach may be adopted for personalized cancer therapy. Since the recent coronavirus pandemic, the development of mRNA vaccines has seen substantial progress and widespread adoption. In the present review, the development of mRNA vaccines, their mechanisms of action, factors influencing their function and the current clinical applications of the vaccine are discussed. A focus is placed on the application of mRNA vaccines in cancer, with the aim of highlighting unique advances and the remaining challenges of this novel and promising therapeutic approach.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251742/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141590309","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that certain of the data shown in Figs. 2A and 4F were strikingly similar to data appearing in different form in other articles written by different authors at different research institutes that were submitted to their respective journals at around the same time; moreover, the same data had apparently been included in the western blots featured in Fig. 5A to show the Parkin and mito‑LCIII protein bands. As it was not clear what had been the original venue for the submission of the strikingly similar data here, the Editor requested that the authors send to us all the raw data underlying the affected figures; however, the authors were not able to comply with this request at the time of asking. Given that the authors were unable to provide the supporting data as requested, the Editor of International Journal of Oncology has decided that this paper should be retracted from the Journal on account of a lack of confidence in the presented data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [International Journal of Oncology 52: 367‑378, 2018; DOI: 10.3892/ijo.2017.4216].
在这篇论文发表后,一位关心此事的读者提请编辑注意,图2A和图4F中显示的某些数据与由不同研究机构的不同作者撰写的其他文章中以不同形式出现的数据惊人地相似,这些文章大约在同一时间提交给了各自的期刊;此外,图5A中的Western印迹显然包含了相同的数据,以显示Parkin和mito-LCIII蛋白条带。由于不清楚这里惊人相似的数据最初是在什么地方提交的,编辑要求作者向我们发送受影响图表的所有原始数据。鉴于作者无法按要求提供支持数据,《国际肿瘤学杂志》编辑决定,由于对所提供的数据缺乏信心,该论文应从杂志上撤下。编辑要求作者解释这些问题,但编辑部没有收到满意的答复。对于给读者带来的不便,编辑深表歉意。[International Journal of Oncology 52: 367-378, 2018; DOI: 10.3892/ijo.2017.4216]。
{"title":"[Retracted] NR4A1‑induced increase in the sensitivity of a human gastric cancer line to TNFα‑mediated apoptosis is associated with the inhibition of JNK/Parkin‑dependent mitophagy.","authors":"Hongzhu Yan, Feng Xiao, Jue Zou, Chengmin Qiu, Weiwei Sun, Minmin Gu, Li Zhang","doi":"10.3892/ijo.2024.5666","DOIUrl":"10.3892/ijo.2024.5666","url":null,"abstract":"<p><p>Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that certain of the data shown in Figs. 2A and 4F were strikingly similar to data appearing in different form in other articles written by different authors at different research institutes that were submitted to their respective journals at around the same time; moreover, the same data had apparently been included in the western blots featured in Fig. 5A to show the Parkin and mito‑LCIII protein bands. As it was not clear what had been the original venue for the submission of the strikingly similar data here, the Editor requested that the authors send to us all the raw data underlying the affected figures; however, the authors were not able to comply with this request at the time of asking. Given that the authors were unable to provide the supporting data as requested, the Editor of <i>International Journal of Oncology</i> has decided that this paper should be retracted from the Journal on account of a lack of confidence in the presented data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [International Journal of Oncology 52: 367‑378, 2018; DOI: 10.3892/ijo.2017.4216].</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 2","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Several studies have indicated that the gut microbiome and tumor microbiota may affect tumors. Emerging metabolomics research illustrates the need to examine the variations in microbial metabolite composition between patients with cancer and healthy individuals. Microbial metabolites can impact the progression of tumors and the immune response by influencing a number of mechanisms, including modulation of the immune system, cancer or immune‑related signaling pathways, epigenetic modification of proteins and DNA damage. Microbial metabolites can also alleviate side effects and drug resistance during chemotherapy and immunotherapy, while effectively activating the immune system to exert tumor immunotherapy. Nevertheless, the impact of microbial metabolites on tumor immunity can be both beneficial and harmful, potentially influenced by the concentration of the metabolites or the specific cancer type. The present review summarizes the roles of various microbial metabolites in different solid tumors, alongside their influence on tumor immunity and treatment. Additionally, clinical trials evaluating the therapeutic effects of microbial metabolites or related microbes on patients with cancer have been listed. In summary, studying microbial metabolites, which play a crucial role in the interaction between the microbiota and tumors, could lead to the identification of new supplementary treatments for cancer. This has the potential to improve the effectiveness of cancer treatment and enhance patient prognosis.
多项研究表明,肠道微生物群和肿瘤微生物群可能会影响肿瘤。新出现的代谢组学研究表明,有必要研究癌症患者和健康人之间微生物代谢物组成的变化。微生物代谢物可通过影响免疫系统、癌症或免疫相关信号通路的调节、蛋白质的表观遗传修饰和 DNA 损伤等多种机制来影响肿瘤的进展和免疫反应。微生物代谢物还能减轻化疗和免疫疗法的副作用和耐药性,同时有效激活免疫系统,发挥肿瘤免疫疗法的作用。然而,微生物代谢物对肿瘤免疫的影响既可能是有益的,也可能是有害的,这可能受到代谢物浓度或特定癌症类型的影响。本综述总结了各种微生物代谢物在不同实体瘤中的作用,以及它们对肿瘤免疫和治疗的影响。此外,还列出了评估微生物代谢物或相关微生物对癌症患者治疗效果的临床试验。总之,微生物代谢物在微生物群与肿瘤之间的相互作用中发挥着至关重要的作用,对微生物代谢物的研究可能有助于确定新的癌症辅助治疗方法。这有可能提高癌症治疗的效果,改善病人的预后。
{"title":"Microbial metabolites affect tumor progression, immunity and therapy prediction by reshaping the tumor microenvironment (Review).","authors":"Yuhang Zhou, Wenjie Han, Yun Feng, Yue Wang, Tao Sun, Junnan Xu","doi":"10.3892/ijo.2024.5661","DOIUrl":"10.3892/ijo.2024.5661","url":null,"abstract":"<p><p>Several studies have indicated that the gut microbiome and tumor microbiota may affect tumors. Emerging metabolomics research illustrates the need to examine the variations in microbial metabolite composition between patients with cancer and healthy individuals. Microbial metabolites can impact the progression of tumors and the immune response by influencing a number of mechanisms, including modulation of the immune system, cancer or immune‑related signaling pathways, epigenetic modification of proteins and DNA damage. Microbial metabolites can also alleviate side effects and drug resistance during chemotherapy and immunotherapy, while effectively activating the immune system to exert tumor immunotherapy. Nevertheless, the impact of microbial metabolites on tumor immunity can be both beneficial and harmful, potentially influenced by the concentration of the metabolites or the specific cancer type. The present review summarizes the roles of various microbial metabolites in different solid tumors, alongside their influence on tumor immunity and treatment. Additionally, clinical trials evaluating the therapeutic effects of microbial metabolites or related microbes on patients with cancer have been listed. In summary, studying microbial metabolites, which play a crucial role in the interaction between the microbiota and tumors, could lead to the identification of new supplementary treatments for cancer. This has the potential to improve the effectiveness of cancer treatment and enhance patient prognosis.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 1","pages":""},"PeriodicalIF":5.2,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11173369/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141283682","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-07-01Epub Date: 2024-05-31DOI: 10.3892/ijo.2024.5658
Tianxiang Zhang, Xi Zhang, Yang Fei, Jinsen Lu, Dairan Zhou, Li Zhang, Song Fan, Jun Zhou, Chaozhao Liang, Yang Su
Clear cell renal cell carcinoma (ccRCC), the most common type of renal cell carcinoma (RCC), is not sensitive to traditional radiotherapy and chemotherapy. The polyphenolic compound Gallic acid (GA) can be naturally found in a variety of fruits, vegetables and plants. Autophagy, an intracellular catabolic process, regulates the lysosomal degradation of organelles and portions in cytoplasm. It was reported that autophagy and GA could affect the development of several cancers. Therefore, the aim of the present study was to evaluate the effects of GA on ccRCC development and clarify the role of autophagy in this process. In the present study, the effects of GA on the proliferation, migration and invasion of ccRCC cells were investigated in vitro by Cell Counting Kit‑8, colony formation, flow cytometry, wound healing and Transwell migration assays, respectively. Additionally, the effects of GA on ccRCC growth and metastasis were evaluated using hematoxylin‑eosin and immunohistochemical staining in vivo. Moreover, it was sought to explore the underlying molecular mechanisms using transmission electron microscopy, western blotting and reverse transcription‑quantitative PCR analyses. In the present study, it was revealed that GA had a more potent viability inhibitory effect on ccRCC cells (786‑O and ACHN) than the effect on normal renal tubular epithelial cell (HK‑2), which demonstrated that GA selectively inhibits the viability of cancer cells. Furthermore, it was identified that GA dose‑dependently inhibited the proliferation, migration and invasion of ccRCC cells in vitro and in vivo. It was demonstrated that GA promoted the release of autophagy markers, which played a role in regulating the PI3K/Akt/Atg16L1 signaling pathway. All the aforementioned data provided evidence for the great potential of GA in the treatment of ccRCC.
透明细胞肾细胞癌(ccRCC)是最常见的肾细胞癌(RCC)类型,对传统的放疗和化疗不敏感。多酚化合物没食子酸(GA)天然存在于各种水果、蔬菜和植物中。自噬是一种细胞内分解代谢过程,调节细胞质中细胞器和部分的溶酶体降解。据报道,自噬和 GA 可影响多种癌症的发展。因此,本研究旨在评估 GA 对 ccRCC 发育的影响,并阐明自噬在这一过程中的作用。本研究通过细胞计数试剂盒-8、菌落形成、流式细胞仪、伤口愈合和 Transwell 迁移试验,分别研究了 GA 对 ccRCC 细胞增殖、迁移和侵袭的影响。此外,还采用苏木精-伊红和免疫组化染色法评估了 GA 对 ccRCC 生长和转移的影响。此外,还利用透射电子显微镜、Western 印迹和逆转录定量 PCR 分析等方法探讨了其潜在的分子机制。本研究发现,与对正常肾小管上皮细胞(HK-2)的抑制作用相比,GA对ccRCC细胞(786-O和ACHN)的活力抑制作用更强,这表明GA能选择性地抑制癌细胞的活力。此外,研究还发现 GA 对体外和体内 ccRCC 细胞的增殖、迁移和侵袭具有剂量依赖性的抑制作用。研究表明,GA 能促进自噬标记物的释放,从而在调节 PI3K/Akt/Atg16L1 信号通路方面发挥作用。所有上述数据都证明了 GA 在治疗 ccRCC 方面的巨大潜力。
{"title":"Gallic acid suppresses the progression of clear cell renal cell carcinoma through inducing autophagy via the PI3K/Akt/Atg16L1 signaling pathway.","authors":"Tianxiang Zhang, Xi Zhang, Yang Fei, Jinsen Lu, Dairan Zhou, Li Zhang, Song Fan, Jun Zhou, Chaozhao Liang, Yang Su","doi":"10.3892/ijo.2024.5658","DOIUrl":"10.3892/ijo.2024.5658","url":null,"abstract":"<p><p>Clear cell renal cell carcinoma (ccRCC), the most common type of renal cell carcinoma (RCC), is not sensitive to traditional radiotherapy and chemotherapy. The polyphenolic compound Gallic acid (GA) can be naturally found in a variety of fruits, vegetables and plants. Autophagy, an intracellular catabolic process, regulates the lysosomal degradation of organelles and portions in cytoplasm. It was reported that autophagy and GA could affect the development of several cancers. Therefore, the aim of the present study was to evaluate the effects of GA on ccRCC development and clarify the role of autophagy in this process. In the present study, the effects of GA on the proliferation, migration and invasion of ccRCC cells were investigated <i>in vitro</i> by Cell Counting Kit‑8, colony formation, flow cytometry, wound healing and Transwell migration assays, respectively. Additionally, the effects of GA on ccRCC growth and metastasis were evaluated using hematoxylin‑eosin and immunohistochemical staining <i>in vivo</i>. Moreover, it was sought to explore the underlying molecular mechanisms using transmission electron microscopy, western blotting and reverse transcription‑quantitative PCR analyses. In the present study, it was revealed that GA had a more potent viability inhibitory effect on ccRCC cells (786‑O and ACHN) than the effect on normal renal tubular epithelial cell (HK‑2), which demonstrated that GA selectively inhibits the viability of cancer cells. Furthermore, it was identified that GA dose‑dependently inhibited the proliferation, migration and invasion of ccRCC cells <i>in vitro</i> and <i>in vivo</i>. It was demonstrated that GA promoted the release of autophagy markers, which played a role in regulating the PI3K/Akt/Atg16L1 signaling pathway. All the aforementioned data provided evidence for the great potential of GA in the treatment of ccRCC.</p>","PeriodicalId":14175,"journal":{"name":"International journal of oncology","volume":"65 1","pages":""},"PeriodicalIF":4.5,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11173374/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141179508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}