{"title":"胎盘间充质干细胞来源的白细胞介素-6促进神经母细胞瘤的进展。","authors":"Ying Liu, Bai Li, Huixia Wei, Yan Xu, Yufeng Liu","doi":"10.62347/KNYX4079","DOIUrl":null,"url":null,"abstract":"<p><p>Neuroblastoma (NB) is a prevalent pediatric malignancy, yet the role of mesenchymal stem cells (MSCs) in NB progression remains unclear. MSCs are known to secrete various cytokines, including interleukin (IL)-6, and their influence on NB cells and tumor xenografts were investigated in this study. Placenta-derived mesenchymal stem cells (PMSCs) were isolated from chorionic villi and characterized via flow cytometry. The obtained PMSCs were co-cultured with NB cells or IL-6-silenced PMSCs. Comprehensive assays were conducted to assess proliferation, colony formation, cell cycle progression, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT). RNA-seq identified differentially expressed genes (DEGs) in NB cells, predominantly enriched in Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways (P < 0.05). qRT-PCR revealed elevated levels of IL-6 and other oncogenic cytokines in PMSCs (P < 0.05). In vivo, IL-6 knockdown in PMSCs significantly suppressed NB xenograft growth, accompanied by reduced expression of Ki-67, proliferating cell nuclear antigen (PCNA), Caspase 9, and Snail as shown by immunohistochemistry (P < 0.05). Western blotting confirmed activation of phosphatidylinositol 3-kinases/protein kinase B (PI3K/AKT) pathway in NB cells after co-culture with PMSCs, which was attenuated by PI3K inhibition. Notably, IL-6 knockdown markedly suppressed NB xenograft progression and downregulated associated signaling markers (P < 0.05). Collectively, PMSC-derived IL-6 potentiates NB progression via PI3K-AKT signaling, presenting a potential therapeutic target in neuroblastoma.</p>","PeriodicalId":7437,"journal":{"name":"American journal of cancer research","volume":"15 8","pages":"3712-3727"},"PeriodicalIF":2.9000,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12432566/pdf/","citationCount":"0","resultStr":"{\"title\":\"Placental mesenchymal stem cell-derived interleukin-6 promotes neuroblastoma progression.\",\"authors\":\"Ying Liu, Bai Li, Huixia Wei, Yan Xu, Yufeng Liu\",\"doi\":\"10.62347/KNYX4079\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Neuroblastoma (NB) is a prevalent pediatric malignancy, yet the role of mesenchymal stem cells (MSCs) in NB progression remains unclear. MSCs are known to secrete various cytokines, including interleukin (IL)-6, and their influence on NB cells and tumor xenografts were investigated in this study. Placenta-derived mesenchymal stem cells (PMSCs) were isolated from chorionic villi and characterized via flow cytometry. The obtained PMSCs were co-cultured with NB cells or IL-6-silenced PMSCs. Comprehensive assays were conducted to assess proliferation, colony formation, cell cycle progression, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT). RNA-seq identified differentially expressed genes (DEGs) in NB cells, predominantly enriched in Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways (P < 0.05). qRT-PCR revealed elevated levels of IL-6 and other oncogenic cytokines in PMSCs (P < 0.05). In vivo, IL-6 knockdown in PMSCs significantly suppressed NB xenograft growth, accompanied by reduced expression of Ki-67, proliferating cell nuclear antigen (PCNA), Caspase 9, and Snail as shown by immunohistochemistry (P < 0.05). Western blotting confirmed activation of phosphatidylinositol 3-kinases/protein kinase B (PI3K/AKT) pathway in NB cells after co-culture with PMSCs, which was attenuated by PI3K inhibition. Notably, IL-6 knockdown markedly suppressed NB xenograft progression and downregulated associated signaling markers (P < 0.05). Collectively, PMSC-derived IL-6 potentiates NB progression via PI3K-AKT signaling, presenting a potential therapeutic target in neuroblastoma.</p>\",\"PeriodicalId\":7437,\"journal\":{\"name\":\"American journal of cancer research\",\"volume\":\"15 8\",\"pages\":\"3712-3727\"},\"PeriodicalIF\":2.9000,\"publicationDate\":\"2025-08-25\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12432566/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"American journal of cancer research\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.62347/KNYX4079\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2025/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q2\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"American journal of cancer research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.62347/KNYX4079","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2025/1/1 0:00:00","PubModel":"eCollection","JCR":"Q2","JCRName":"ONCOLOGY","Score":null,"Total":0}
Neuroblastoma (NB) is a prevalent pediatric malignancy, yet the role of mesenchymal stem cells (MSCs) in NB progression remains unclear. MSCs are known to secrete various cytokines, including interleukin (IL)-6, and their influence on NB cells and tumor xenografts were investigated in this study. Placenta-derived mesenchymal stem cells (PMSCs) were isolated from chorionic villi and characterized via flow cytometry. The obtained PMSCs were co-cultured with NB cells or IL-6-silenced PMSCs. Comprehensive assays were conducted to assess proliferation, colony formation, cell cycle progression, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT). RNA-seq identified differentially expressed genes (DEGs) in NB cells, predominantly enriched in Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways (P < 0.05). qRT-PCR revealed elevated levels of IL-6 and other oncogenic cytokines in PMSCs (P < 0.05). In vivo, IL-6 knockdown in PMSCs significantly suppressed NB xenograft growth, accompanied by reduced expression of Ki-67, proliferating cell nuclear antigen (PCNA), Caspase 9, and Snail as shown by immunohistochemistry (P < 0.05). Western blotting confirmed activation of phosphatidylinositol 3-kinases/protein kinase B (PI3K/AKT) pathway in NB cells after co-culture with PMSCs, which was attenuated by PI3K inhibition. Notably, IL-6 knockdown markedly suppressed NB xenograft progression and downregulated associated signaling markers (P < 0.05). Collectively, PMSC-derived IL-6 potentiates NB progression via PI3K-AKT signaling, presenting a potential therapeutic target in neuroblastoma.
期刊介绍:
The American Journal of Cancer Research (AJCR) (ISSN 2156-6976), is an independent open access, online only journal to facilitate rapid dissemination of novel discoveries in basic science and treatment of cancer. It was founded by a group of scientists for cancer research and clinical academic oncologists from around the world, who are devoted to the promotion and advancement of our understanding of the cancer and its treatment. The scope of AJCR is intended to encompass that of multi-disciplinary researchers from any scientific discipline where the primary focus of the research is to increase and integrate knowledge about etiology and molecular mechanisms of carcinogenesis with the ultimate aim of advancing the cure and prevention of this increasingly devastating disease. To achieve these aims AJCR will publish review articles, original articles and new techniques in cancer research and therapy. It will also publish hypothesis, case reports and letter to the editor. Unlike most other open access online journals, AJCR will keep most of the traditional features of paper print that we are all familiar with, such as continuous volume, issue numbers, as well as continuous page numbers to retain our comfortable familiarity towards an academic journal.