Cellular Oncology最新文献

筛选
英文 中文
Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. 肿瘤浸润性骨髓细胞;肿瘤治疗的机制、功能意义和靶向性。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-06-01 Epub Date: 2025-02-25 DOI: 10.1007/s13402-025-01051-y
Fatemeh Sadat Toghraie, Maryam Bayat, Mahsa Sadat Hosseini, Amin Ramezani
{"title":"Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy.","authors":"Fatemeh Sadat Toghraie, Maryam Bayat, Mahsa Sadat Hosseini, Amin Ramezani","doi":"10.1007/s13402-025-01051-y","DOIUrl":"10.1007/s13402-025-01051-y","url":null,"abstract":"<p><p>Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"559-590"},"PeriodicalIF":4.9,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12119771/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143494448","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Tumor-specific CXCR6 positive precursor CD8+ T cells mediate tumor control in metastatic melanoma. 肿瘤特异性CXCR6阳性前体CD8+ T细胞介导转移性黑色素瘤的肿瘤控制。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-06-01 Epub Date: 2025-04-07 DOI: 10.1007/s13402-025-01040-1
Yang Song, Ji Chen, Yaqin Zhang, Ning Wu, Yongjun Zhu, Gang Chen, Feng Miao, Zhiming Chen, Yiqing Wang
{"title":"Tumor-specific CXCR6 positive precursor CD8<sup>+</sup> T cells mediate tumor control in metastatic melanoma.","authors":"Yang Song, Ji Chen, Yaqin Zhang, Ning Wu, Yongjun Zhu, Gang Chen, Feng Miao, Zhiming Chen, Yiqing Wang","doi":"10.1007/s13402-025-01040-1","DOIUrl":"10.1007/s13402-025-01040-1","url":null,"abstract":"<p><strong>Background: </strong>Adoptive cell therapy (ACT) mediates durable and complete regression of various cancers. However, its efficacy is limited by the long-term persistence of cytotoxic T lymphocytes, given their irreversible dysfunction within the tumor microenvironment. Herein, we aimed to establish an artificial lung metastasis model to examine T-lymphocyte subsets, in order to identify potential effective cell subsets for ACT.</p><p><strong>Methods: </strong>A metastatic lung melanoma mouse model was established using OVA-expressing melanoma B16 cells. Flow cytometry analysis was conducted to examine the surface markers, transcription factors, and secreted cytokines of tumor-specific CD8<sup>+</sup> T cells within metastatic tissues. The infiltrated cells were sorted by flow cytometry for in vitro tumor cell killing assays or in vivo cell infusion therapy combined with chemotherapeutic drugs and immune checkpoint blockade antibodies.</p><p><strong>Results: </strong>Exhausted CD8<sup>+</sup> T cells (Tex) exhibited high heterogeneity in metastatic tissues. Among Tex cells, the CXCR6<sup>-</sup> precursor cell showed certain memory characteristics, including phenotype, transcription factors, and maintenance, whereas the CXCR6<sup>+</sup> subpopulation partially lost these traits. Moreover, CXCR6<sup>+</sup> precursor cells effectively replenished effector-like Tex cells in metastatic tissues and exerted direct cytotoxicity against tumor cells. Notably, transferring these tumor-specific CXCR6<sup>+</sup> precursor-exhausted T (Texp) cells into recipients induced a substantial regression of metastasis. In addition, these cells could respond to immune checkpoint blockade, which could better control tumor metastasis.</p><p><strong>Conclusions: </strong>In our study, a subset of antigen-specific CXCR6-expressing Texp cells was observed within the metastatic tissue. The cells served as a crucial source of effector-like Tex cells and exerted direct cytotoxic effects on tumor cells. Adoptive transfer of CXCR6<sup>+</sup> Texp cells effectively mitigated lung metastasis in mice. This study helps elucidate the role of Texp cells in metastasis, thereby offering novel insights into enhancing the efficacy and durability of immunotherapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"693-708"},"PeriodicalIF":4.9,"publicationDate":"2025-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12119687/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143796855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Senolytics: charting a new course or enhancing existing anti-tumor therapies? 老年学:制定新的治疗方案还是加强现有的抗肿瘤疗法?
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-04-01 Epub Date: 2024-12-04 DOI: 10.1007/s13402-024-01018-5
Konrad Czajkowski, Mariola Herbet, Marek Murias, Iwona Piątkowska-Chmiel
{"title":"Senolytics: charting a new course or enhancing existing anti-tumor therapies?","authors":"Konrad Czajkowski, Mariola Herbet, Marek Murias, Iwona Piątkowska-Chmiel","doi":"10.1007/s13402-024-01018-5","DOIUrl":"10.1007/s13402-024-01018-5","url":null,"abstract":"<p><p>Cell senescence is a natural response within our organisms. Initially, it was considered an effective anti-tumor mechanism. However, it is now believed that while cell senescence initially acts as a robust barrier against tumor initiation, the subsequent accumulation of senescent cells can paradoxically promote cancer recurrence and cause damage to neighboring tissues. This intricate balance between cell proliferation and senescence plays a pivotal role in maintaining tissue homeostasis. Moreover, senescence cells secrete many bioactive molecules collectively termed the senescence-associated secretory phenotype (SASP), which can induce chronic inflammation, alter tissue architecture, and promote tumorigenesis through paracrine signaling. Among the myriads of compounds, senotherapeutic drugs have emerged as exceptionally promising candidates in anticancer treatment. Their ability to selectively target senescent cells while sparing healthy tissues represents a paradigm shift in therapeutic intervention, offering new avenues for personalized oncology medicine. Senolytics have introduced new therapeutic possibilities by enabling the targeted removal of senescent cells. As standalone agents, they can clear tumor cells in a senescent state and, when combined with chemo- or radiotherapy, eliminate residual senescent cancer cells after treatment. This dual approach allows for the intentional use of lower-dose therapies or the removal of unintended senescent cells post-treatment. Additionally, by targeting non-cancerous senescent cells, senolytics may help reduce tumor formation risk, limit recurrence, and slow disease progression. This article examines the mechanisms of cellular senescence, its role in cancer treatment, and the importance of senotherapy, with particular attention to the therapeutic potential of senolytic drugs.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"351-371"},"PeriodicalIF":4.9,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11996976/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142781643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Regulatory factor X1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by transcriptional regulation of BECN1. 调节因子X1通过BECN1的转录调控促进索拉非尼诱导的肝癌铁下垂。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-04-01 Epub Date: 2024-12-09 DOI: 10.1007/s13402-024-01017-6
Zhiwen Yang, Yichuan Yuan, Yi Niu, Dinglan Zuo, Wenwu Liu, Kai Li, Yunxing Shi, Zhiyu Qiu, Keren Li, Zhu Lin, Chengrui Zhong, Zhenkun Huang, Wei He, Xinyuan Guan, Yunfei Yuan, Weian Zeng, Jiliang Qiu, Binkui Li
{"title":"Regulatory factor X1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by transcriptional regulation of BECN1.","authors":"Zhiwen Yang, Yichuan Yuan, Yi Niu, Dinglan Zuo, Wenwu Liu, Kai Li, Yunxing Shi, Zhiyu Qiu, Keren Li, Zhu Lin, Chengrui Zhong, Zhenkun Huang, Wei He, Xinyuan Guan, Yunfei Yuan, Weian Zeng, Jiliang Qiu, Binkui Li","doi":"10.1007/s13402-024-01017-6","DOIUrl":"10.1007/s13402-024-01017-6","url":null,"abstract":"<p><strong>Background: </strong>Sorafenib is a commonly used first-line kinase-targeted drug for advanced hepatocellular carcinoma (HCC) patients suffering from limited efficacy. Emerging evidence indicates that sorafenib exerts anti-cancer activity through the induction of ferroptosis in HCC cells, but the underlying mechanism is still unclear.</p><p><strong>Methods: </strong>The whole transcriptome sequencing and bioinformatics analysis were used to screen for target genes. The expression and subcellular localization of regulatory factor X1 (RFX1) were determined through immunohistochemistry, immunofluorescence, PCR and western blot analyses. The impact of RFX1 on HCC cell growth was assessed using CCK8, colony formation assays, cell death assays, and animal experiments. Glutathione measurement, iron assay and lipid peroxidation detection assays were performed to investigate ferroptosis of HCC cells. The regulatory mechanism of RFX1 in HCC was investigated by sgRFX1, co-IP, ChIP and luciferase experiments. Immunohistochemical and survival analyses were performed to examine the prognostic significance of RFX1 in HCC.</p><p><strong>Results: </strong>In this study, we found that RFX1 promote ferroptosis in HCC cells. Further, we showed that sorafenib induces cell death through RFX1-mediated ferroptosis in HCC cells. The enhancing effect of RFX1 on HCC cell ferroptosis is largely dependent on inhibition of cystine/glutamate antiporter (system Xc-) activity through the BECN-SLC7A11 axis, where RFX1 directly binds to the promoter region of BECN1 and upregulates BECN1 expression. In addition, a STAT3-RFX1-BECN1 signalling loop was found to promote RFX1 expression in HCC cells.</p><p><strong>Conclusions: </strong>Our study reveals a novel mechanism underlying sorafenib-induced HCC cell death.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"505-522"},"PeriodicalIF":4.9,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11996997/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142802480","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. 揭示 Th 亚群:深入了解它们在免疫检查点抑制剂疗法中的作用。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-04-01 Epub Date: 2024-09-26 DOI: 10.1007/s13402-024-00992-0
Monika Ryba-Stanisławowska
{"title":"Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy.","authors":"Monika Ryba-Stanisławowska","doi":"10.1007/s13402-024-00992-0","DOIUrl":"10.1007/s13402-024-00992-0","url":null,"abstract":"<p><p>T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"295-312"},"PeriodicalIF":4.9,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11996958/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142330863","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Lenvatinib-activated NDUFA4L2/IL33/PADI4 pathway induces neutrophil extracellular traps that inhibit cuproptosis in hepatocellular carcinoma. 伦伐替尼激活的NDUFA4L2/IL33/PADI4通路可诱导中性粒细胞胞外陷阱,从而抑制肝细胞癌的杯突症。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-04-01 Epub Date: 2024-11-25 DOI: 10.1007/s13402-024-01013-w
Nan Yi, Lingyun Zhang, Xiangbo Huang, Jilei Ma, Jian Gao
{"title":"Lenvatinib-activated NDUFA4L2/IL33/PADI4 pathway induces neutrophil extracellular traps that inhibit cuproptosis in hepatocellular carcinoma.","authors":"Nan Yi, Lingyun Zhang, Xiangbo Huang, Jilei Ma, Jian Gao","doi":"10.1007/s13402-024-01013-w","DOIUrl":"10.1007/s13402-024-01013-w","url":null,"abstract":"<p><strong>Background: </strong>Lenvatinib is a potent first-line therapy for patients with hepatocellular carcinoma (HCC), but it also increased the number of neutrophils in HCC tumor microenvironment.</p><p><strong>Methods: </strong>CitH3, MPO-DNA, elastase and MPO activity were measured for assessing neutrophil extracellular traps (NETs) in vivo and in vitro. Cell cuproptosis was assessed by measurement of copper content, FDX1, and pyruvate. The functions of lenvatinib, DNase I, interleukin 33 (IL33) neutralizing antibody and GPX4 in tumor growth were explored in mice.</p><p><strong>Results: </strong>Lenvatinib induced NETs in the HCC tumor microenvironment via HCC cells, but not through the direct stimulation of neutrophils. In addition, NET clearance by DNase I improves the efficacy of lenvatinib therapy in HCC mouse models. Mechanistically, lenvatinib promoted the expression and secretion of IL33 by HCC cells that triggered NET formation. Moreover, IL33 knockdown in Hepa1-6 cells improved lenvatinib efficacy in Hepa1-6-bearing HCC model mice and reduced NET formation in the tumor microenvironment. Subsequently, lenvatinib increased IL33 production by increasing the NDUFA4L2 expression in HCC cells. Furthermore, we found that IL33 triggered NET formation in neutrophils by increasing the protein expression of PADI4 via the Akt/mTOR signaling pathway. Rapamycin inhibition of mTOR reduced PADI4 expression and NET formation. Consistently, PADI4 inhibition by the selective PAD4 inhibitor GSK484 hydrochloride (GSK484) improved lenvatinib response to HCC therapy. Importantly, NETs contribute to lenvatinib resistance by inhibiting cuproptosis, but not apoptosis, pyroptosis, or ferroptosis in HCC cells. Treatment with GSK484 reversed the inhibitory effects of NETs on cuproptosis and sensitized the HCC cells to lenvatinib.</p><p><strong>Conclusions: </strong>Our study revealed that lenvatinib-induced NETs inhibited the cuproptosis of HCC cells, suggesting that targeting the IL33/PADI4/NET axis represents a promising therapeutic strategy for ameliorating lenvatinib resistance in HCC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"487-504"},"PeriodicalIF":4.9,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11996955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142711554","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
TRPV4 drives the progression of leiomyosarcoma by promoting ECM1 generation and co-activating the FAK/PI3K/AKT/GSK3β pathway. TRPV4通过促进ECM1的生成和共同激活FAK/PI3K/AKT/GSK3β通路来驱动平滑肌肉瘤的进展。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-04-01 Epub Date: 2024-11-29 DOI: 10.1007/s13402-024-01008-7
Qiwen Zhou, Yang You, Yingying Zhao, Shuxiu Xiao, Zhengqing Song, Chuxin Huang, Jiali Qian, Weiqi Lu, Hanxing Tong, Yong Zhang, Zhiming Wang, Wei Li, Chenlu Zhang, Xi Guo, Rongkui Luo, Yingyong Hou, Jiefeng Cui, Lili Lu, Yuhong Zhou
{"title":"TRPV4 drives the progression of leiomyosarcoma by promoting ECM1 generation and co-activating the FAK/PI3K/AKT/GSK3β pathway.","authors":"Qiwen Zhou, Yang You, Yingying Zhao, Shuxiu Xiao, Zhengqing Song, Chuxin Huang, Jiali Qian, Weiqi Lu, Hanxing Tong, Yong Zhang, Zhiming Wang, Wei Li, Chenlu Zhang, Xi Guo, Rongkui Luo, Yingyong Hou, Jiefeng Cui, Lili Lu, Yuhong Zhou","doi":"10.1007/s13402-024-01008-7","DOIUrl":"10.1007/s13402-024-01008-7","url":null,"abstract":"<p><strong>Purpose: </strong>Leiomyosarcoma (LMS) is an aggressive mesenchymal malignant tumor with poor therapeutic options, but the molecular mechanisms underlying LMS remain largely unknown. Increasing evidence indicates that transient receptor potential vanilloid 4 (TRPV4) levels are closely related to the advancement of various malignant tumors through diverse molecular mechanisms. However, the roles and regulatory mechanisms of TRPV4 in LMS progression remain unclear.</p><p><strong>Methods: </strong>Immunohistochemistry, Western blot, and immunofluorescence were used to investigate the relationship between TRPV4 expression and LMS. Survival analysis was conducted to evaluate the association between TRPV4 levels and prognosis in LMS patients. Intracellular Ca<sup>2+</sup> measurement, colony formation, CCK-8, wound healing and Transwell assays and peritoneal metastasis mouse model were used to verify the effect of TRPV4 activity and expression on LMS proliferation and metastasis. RNA-seq and proteomics were performed to explore the underlying mechanism.</p><p><strong>Results: </strong>TRPV4 was upregulated in LMS tissues and cells and served as a novel prognostic factor. Moreover, TRPV4 overexpression enhanced cell proliferation, cell migration and invasion of LMS cells in vitro, as well as promoted tumor metastasis in vivo, which could be blocked by HC067047 intervention or TRPV4 knockdown. Combined RNA-seq and proteomics analysis of KEGG pathway indicated that ECM receptor interaction was obviously activated. Extracellular matrix protein 1 (ECM1) was identified as downstream gene of TRPV4. Mechanistically, TRPV4 overexpression increased ECM1 level and activated the FAK/PI3K/AKT/GSK3β pathway, which could be reversed by TRPV4 knockdown or LY294002 treatment. Moreover, ECM1 overexpression enhanced the activation of FAK/PI3K/AKT/GSK3β pathway. And simultaneous overexpression of TRPV4 and ECM1 synergistically activated this pathway.</p><p><strong>Conclusion: </strong>Our findings provide a novel mechanism by which TRPV4 directly activates Ca<sup>2+</sup>/FAK/PI3K/AKT/GSK3β pathway and further indirectly enhances the FAK/PI3K/AKT/GSK3β pathway through the promotion and secretion of ECM1 to promote LMS malignant progression. Targeting the TRPV4/FAK axis might be a promising potential strategy for prognosis and treatment of LMS.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"455-470"},"PeriodicalIF":4.9,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11996984/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142752115","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Comprehensive multiomics analysis of the signatures of gastric mucosal bacteria and plasma metabolites across different stomach microhabitats in the development of gastric cancer. 对胃癌发生过程中不同胃微生境下胃黏膜细菌和血浆代谢物特征的多组学综合分析
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-02-01 Epub Date: 2024-07-04 DOI: 10.1007/s13402-024-00965-3
Bingsen Wang, Jiahui Luan, Weidong Zhao, Junbao Yu, Anqing Li, Xinxin Li, Xiaoqin Zhong, Hongyun Cao, Ruicai Wang, Bo Liu, Shiyong Lu, Mei Shi
{"title":"Comprehensive multiomics analysis of the signatures of gastric mucosal bacteria and plasma metabolites across different stomach microhabitats in the development of gastric cancer.","authors":"Bingsen Wang, Jiahui Luan, Weidong Zhao, Junbao Yu, Anqing Li, Xinxin Li, Xiaoqin Zhong, Hongyun Cao, Ruicai Wang, Bo Liu, Shiyong Lu, Mei Shi","doi":"10.1007/s13402-024-00965-3","DOIUrl":"10.1007/s13402-024-00965-3","url":null,"abstract":"<p><strong>Purpose: </strong>As an important component of the microenvironment, the gastric microbiota and its metabolites are associated with tumour occurrence, progression, and metastasis. However, the relationship between the gastric microbiota and the development of gastric cancer is unclear. The present study investigated the role of the gastric mucosa microbiome and metabolites as aetiological factors in gastric carcinogenesis.</p><p><strong>Methods: </strong>Gastric biopsies from different stomach microhabitats (n = 70) were subjected to 16S rRNA gene sequencing, and blood samples (n = 95) were subjected to untargeted metabolome (gas chromatography‒mass spectrometry, GC‒MS) analyses. The datasets were analysed using various bioinformatics approaches.</p><p><strong>Results: </strong>The microbiota diversity and community composition markedly changed during gastric carcinogenesis. High Helicobacter. pylori colonization modified the overall diversity and composition of the microbiota associated with gastritis and cancer in the stomach. Most importantly, analysis of the functional features of the microbiota revealed that nitrate reductase genes were significantly enriched in the tumoral microbiota, while urease-producing genes were significantly enriched in the microbiota of H. pylori-positive patients. A panel of 81 metabolites was constructed to discriminate gastric cancer patients from gastritis patients, and a panel of 15 metabolites was constructed to discriminate H. pylori-positive patients from H. pylori-negative patients. receiver operator characteristic (ROC) curve analysis identified a series of gastric microbes and plasma metabolites as potential biomarkers of gastric cancer.</p><p><strong>Conclusion: </strong>The present study identified a series of signatures that may play important roles in gastric carcinogenesis and have the potential to be used as biomarkers for diagnosis and for the surveillance of gastric cancer patients with minimal invasiveness.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"139-159"},"PeriodicalIF":4.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850404/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141499417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. 三维肿瘤细胞培养方法及在癌症研究和个性化医疗中的应用系统综述。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-02-01 Epub Date: 2024-05-28 DOI: 10.1007/s13402-024-00960-8
Jessica Kalla, Janette Pfneissl, Theresia Mair, Loan Tran, Gerda Egger
{"title":"A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine.","authors":"Jessica Kalla, Janette Pfneissl, Theresia Mair, Loan Tran, Gerda Egger","doi":"10.1007/s13402-024-00960-8","DOIUrl":"10.1007/s13402-024-00960-8","url":null,"abstract":"<p><p>Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1-26"},"PeriodicalIF":4.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850459/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141161474","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Telomere transcripts act as tumor suppressor and are associated with favorable prognosis in colorectal cancer with low proliferating cell nuclear antigen expression. 端粒转录物是肿瘤抑制因子,与增殖细胞核抗原低表达的结直肠癌的良好预后有关。
IF 4.9 2区 医学
Cellular Oncology Pub Date : 2025-02-01 Epub Date: 2024-09-02 DOI: 10.1007/s13402-024-00986-y
Philip Kienzl, Abigail J Deloria, Monika Hunjadi, Juliane M Hadolt, Max-Felix Haering, Angrit Bothien, Doris Mejri, Medina Korkut-Demirbaş, Sandra Sampl, Gerhard Weber, Christine Pirker, Severin Laengle, Tamara Braunschmid, Eleni Dragona, Brigitte Marian, Sarantis Gagos, Lingeng Lu, Jeremy D Henson, Loretta M S Lau, Roger R Reddel, Wolfgang Mikulits, Stefan Stättner, Klaus Holzmann
{"title":"Telomere transcripts act as tumor suppressor and are associated with favorable prognosis in colorectal cancer with low proliferating cell nuclear antigen expression.","authors":"Philip Kienzl, Abigail J Deloria, Monika Hunjadi, Juliane M Hadolt, Max-Felix Haering, Angrit Bothien, Doris Mejri, Medina Korkut-Demirbaş, Sandra Sampl, Gerhard Weber, Christine Pirker, Severin Laengle, Tamara Braunschmid, Eleni Dragona, Brigitte Marian, Sarantis Gagos, Lingeng Lu, Jeremy D Henson, Loretta M S Lau, Roger R Reddel, Wolfgang Mikulits, Stefan Stättner, Klaus Holzmann","doi":"10.1007/s13402-024-00986-y","DOIUrl":"10.1007/s13402-024-00986-y","url":null,"abstract":"<p><p>Telomeric repeat-containing RNAs (TERRA) and telomerase RNA component (TERC) regulate telomerase activity (TA) and thereby contribute to telomere homeostasis by influencing telomere length (TL) and the cell immortality hallmark of cancer cells. Additionally, the non-canonical functions of telomerase reverse transcriptase (TERT) and TERRA appear to be involved in the epithelial-mesenchymal transition (EMT), which is important for cancer progression. However, the relationship between TERRA and patient prognosis has not been fully characterized. In this small-scale study, 68 patients with colorectal cancer (CRC) were evaluated for correlations between telomere biology, proliferation, and EMT gene transcripts and disease outcome. The proliferating cell nuclear antigen (PCNA) and the epithelial splicing regulatory proteins 1 and 2 (ESRP1 and ESRP2) showed a positive correlation with TERRA, while TA and TERRA exhibited an inverse correlation. Consistent with previous findings, the present study revealed higher expression levels of TERT and TERC, and increased TA and TL in CRC tumor tissue compared to adjacent non-tumor tissue. In contrast, lower expression levels of TERRA were observed in tumor tissue. Patients with high TERRA expression and low PCNA levels exhibited favorable overall survival rates compared to individuals with the inverse pattern. Furthermore, TERRA suppressed CRC tumor growth in severe combined immunodeficiency disease (SCID) mice. In conclusion, our study extends previously published research on TERRA suggesting its potential therapeutic role in telomerase-positive CRC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"239-247"},"PeriodicalIF":4.9,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850466/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142113993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信