Cancer Cell International最新文献

筛选
英文 中文
A Mendelian randomization study of type 2 diabetes and cancer risk in East Asians. 东亚2型糖尿病和癌症风险的孟德尔随机研究。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-08-04 DOI: 10.1186/s12935-025-03929-1
Ling Li, Fangxuan Li, Zhanyu Pan
{"title":"A Mendelian randomization study of type 2 diabetes and cancer risk in East Asians.","authors":"Ling Li, Fangxuan Li, Zhanyu Pan","doi":"10.1186/s12935-025-03929-1","DOIUrl":"10.1186/s12935-025-03929-1","url":null,"abstract":"<p><p>Our research aims to explore genetic correlation between T2D predisposition and risks of several cancers, which have been predominantly focused on populations of European ancestry. In an East Asian population, we leverage two-sample Mendelian Randomization to investigate the complex association between Type 2 Diabetes (T2D) and cancer susceptibility. This investigation utilizes genetic data summarized from three reputable sources: the Japanese ENcyclopedia of GEnetic associations by Riken (JENGER), the Asian Genetic Epidemiology Network (AGEN), and the Meta Analyses of Glucose and Insulin-related traits (MAGIC). We explored the associations between exposure datasets, which included T2D, glycated hemoglobin (HbA1c) and fasting glucose (FG) levels, and the risk of several prevalent cancers for the outcome datasets. By analyzing 174 SNPs associated with T2D, 15 SNPs related to FG, and 74 SNPs linked to HbA1c, we discovered a significant inverse relationship between T2D and the majority of cancers, including gastric (OR = 0.875, 95%CI: 0.825-0.928), breast (OR = 0.907, 95%CI: 0.850-0.967), esophageal (OR = 0.761, 95%CI:0.681-0.851), colorectal (OR = 0.877, 95%CI:0.834-0.923), hematological malignancy (OR = 0.837, 95%CI:0.752-0.933), lung (OR = 0.916, 95%CI:0.858-0.977), hepatocellular (OR = 0.865, 95%CI:0.787-0.951), prostate (OR = 0.862, 95%CI:0.812-0.914), and endometrial cancer (OR = 0.841). The power of gastric cancer, breast cancer, esophageal cancer, colorectal cancer and prostate cancer reached the desired 80%. Conversely, we did not observe a reverse correlation between T2D and specific cancers in the East Asian population. There is limited evidence that genetically predicted FG and HbA1c levels are associated with specific cancer. Moreover, conducting multivariable MR analysis did not alter the correlation between T2D and specific tumors. These findings explain the causal associations of genetic liability to T2D with various types of cancers in East Asian ancestry, and this association is not affected by glycemic biomarkers.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"295"},"PeriodicalIF":6.0,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12320361/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144783571","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
Periplocin potentiates ferroptotic cell death in non-small cell lung cancer by inducing the degradation of Nrf2. Periplocin通过诱导Nrf2的降解加速非小细胞肺癌的铁致细胞死亡。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-08-04 DOI: 10.1186/s12935-025-03925-5
Jinhao Wang, Yue Zhu, Jialiang Song, Fengping Yang, Peng Wang, Ziyi Zhang, Yuxuan Li, Minghe Dai, Yinuo Wang, Waleed Yousuf, Jiayu Li, Dian Yang, Shaoxuan Cheng, Shuyan Liu, Zhaoxia Dai, Xun Qiu, Yingqiu Zhang, Zengchun Hu
{"title":"Periplocin potentiates ferroptotic cell death in non-small cell lung cancer by inducing the degradation of Nrf2.","authors":"Jinhao Wang, Yue Zhu, Jialiang Song, Fengping Yang, Peng Wang, Ziyi Zhang, Yuxuan Li, Minghe Dai, Yinuo Wang, Waleed Yousuf, Jiayu Li, Dian Yang, Shaoxuan Cheng, Shuyan Liu, Zhaoxia Dai, Xun Qiu, Yingqiu Zhang, Zengchun Hu","doi":"10.1186/s12935-025-03925-5","DOIUrl":"10.1186/s12935-025-03925-5","url":null,"abstract":"<p><strong>Background: </strong>Non-small cell lung cancer represents the main histological subtype of lung cancer. Periplocin is a major cardiac glycoside found in the traditional Chinese medicine Cortex periplocae administered in cardiovascular and autoimmune diseases. Inspired by recent findings reporting the anticancer activities of periplocin, this study investigates its potential effects against lung cancer.</p><p><strong>Methods: </strong>The influence of periplocin on non-small cell lung cancer cells was examined by CCK-8, colony formation, and EdU staining assays, followed by transcriptomic profiling with RNA sequencing. Gene set enrichment analysis was conducted to identify pathways affected by periplocin. Nrf2 expression was assessed by Western blotting and turnover was investigated by cycloheximide chase assays. Cellular ferroptosis was induced by the GPX4 inhibitor with or without periplocin treatment. The in vivo effects of periplocin were assessed using lung cancer xenograft mouse models.</p><p><strong>Results: </strong>Periplocin inhibited lung cancer cell growth in vitro. Transcriptomic analysis showed significant downregulation of Nrf2 downstream targets. Biochemical characterization revealed that periplocin increased Nrf2 turnover by promoting proteasomal degradation, leading to decreased levels of downstream transcripts. Functionally, Nrf2 reduction imposed by periplocin treatment rendered lung cancer cells increased susceptibility to ferroptosis induction. Finally, periplocin treatment demonstrated similar inhibition to restrict lung cancer xenograft growth as compared to the ferroptosis inducer imidazole ketone erastin, with both compounds leading to elevated expression of the ferroptosis marker COX2 in xenograft tumor tissues.</p><p><strong>Conclusion: </strong>Our investigation suggests periplocin as a potential agent in the development of ferroptosis-inducing therapies against non-small cell lung cancer.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"294"},"PeriodicalIF":6.0,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12320301/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144783572","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
Understanding crosstalk between the gut and liver microbiome: pathogenesis to therapeutic approaches in liver cancer. 了解肠道和肝脏微生物组之间的串扰:肝癌的发病机制和治疗方法。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-29 DOI: 10.1186/s12935-025-03840-9
Himanshi Goyal, Kaneez Fatima, Jyotdeep Kaur
{"title":"Understanding crosstalk between the gut and liver microbiome: pathogenesis to therapeutic approaches in liver cancer.","authors":"Himanshi Goyal, Kaneez Fatima, Jyotdeep Kaur","doi":"10.1186/s12935-025-03840-9","DOIUrl":"10.1186/s12935-025-03840-9","url":null,"abstract":"<p><p>HCC is the most common primary liver cancer, ranking as the sixth most prevalent cancer globally and the third leading cause of cancer-related deaths. Etiological factors include:chronic liver diseases driven by alcohol abuse, viral hepatitis, obesity, and metabolic disorders. Emerging evidence also suggests that gut microbiome alterations and subsequent immune and metabolic dysregulation contribute to the pathogenesis and progression of HCC. The gut-liver axis represents a dynamic interplay between the gastrointestinal tract and the liver, modulated by the gut microbiome, microbial metabolites, and immune responses. This bidirectional communication plays a pivotal role in maintaining metabolic homeostasis and immune surveillance, while its dysregulation is implicated in various pathologies, including hepatocellular carcinoma (HCC). The gut microbiome, through microbial dysbiosis and metabolite secretion, significantly influences the tumor microenvironment and immune evasion mechanisms in HCC. Perturbations in gut barrier function and Toll-like receptor 4 (TLR4) activation drive chronic inflammation, promoting tumor progression. Moreover, microbial metabolites such as short-chain fatty acids (SCFAs) and bile acids, modulate inflammatory and metabolic pathways, offering novel insights into disease pathogenesis and potential biomarkers. Therapeutic strategies, including probiotics, prebiotics, and immune checkpoint inhibitors demonstrate promise in reprogramming the gut microbiome and restoring immune balance in HCC management. This review explores the multifaceted roles of the gut-liver axis in pathogenesis, the contributions of the intra-tumoral microbiome, and the potential of microbial metabolites as therapeutic avenues. A deeper understanding of these interactions could pave the way for innovative, targeted interventions in liver cancer and other gut-liver axis-associated diseases.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"291"},"PeriodicalIF":6.0,"publicationDate":"2025-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12305952/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741275","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
Targeting DKC1/NF-κB axis suppresses tumorigenesis and enhances 5-FU sensitivity in gastric cancer. 靶向DKC1/NF-κB轴抑制胃癌发生,增强5-FU敏感性。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03926-4
Tengkai Wang, Hui Zhang, Yaoyao Feng, Yinrong Yang
{"title":"Targeting DKC1/NF-κB axis suppresses tumorigenesis and enhances 5-FU sensitivity in gastric cancer.","authors":"Tengkai Wang, Hui Zhang, Yaoyao Feng, Yinrong Yang","doi":"10.1186/s12935-025-03926-4","DOIUrl":"10.1186/s12935-025-03926-4","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC), an aggressive malignant tumor with poor overall survival worldwide, urgently requires novel diagnostic and therapeutic targets. The dyskeratosis congenita 1 (DKC1) gene has been reported to have diverse biological functions and prognostic values in several types of human cancers. However, the specific role and molecular mechanism of DKC1 in GC have received less attention.</p><p><strong>Methods: </strong>Multi-omics analysis integrated The Cancer Genome Atlas (TCGA) data with validation in GC specimens and GC cell lines. DKC1 expression was quantified via immunohistochemistry, real-time polymerase chain reaction (qRT-PCR), and western blotting. Functional impacts on proliferation (CCK-8/cell cycle analysis), migration (Transwell), apoptosis (Annexin V/PI staining), and chemosensitivity (5-fluorouracil [5-FU] IC50) were assessed. RNA sequencing of DKC1-silenced AGS cells informed pathway enrichment (Gene Set Enrichment Analysis [GSEA]/Kyoto Encyclopedia of Genes and Genomes [KEGG]) to predict the underlying mechanism.</p><p><strong>Results: </strong>DKC1 expression was increased and displayed a remarkable diagnostic value in GC. High DKC1 expression correlated with advanced histologic grade and diffuse-type Lauren classification in GC patients. Functional studies revealed that DKC1 effectively promoted GC cell proliferation and migration while suppressing apoptosis in vitro. RNA-seq analysis and rescue experiments confirmed that DKC1 regulated GC progression via the NF-κB signaling pathway. Critically, DKC1 knockdown synergistically enhanced 5-FU efficacy through cell cycle dysregulation.</p><p><strong>Conclusions: </strong>DKC1 was identified as a regulator of GC development through the NF-κB pathway. It displayed a dual role as a diagnostic biomarker and therapeutic target. Elevated DKC1 expression correlated with aggressive clinicopathological features and a good diagnostic value in GC. Furthermore, DKC1 knockdown synergistically enhanced 5-FU efficacy. These data suggested that DKC1 is a potential tumor diagnostic biomarker and a therapeutic target for GC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"288"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12302887/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728098","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
Targeting cancer stem cells with CAR-based immunotherapy: biology, evidence, and future directions. 靶向肿瘤干细胞的CAR-based免疫疗法:生物学、证据和未来方向。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03846-3
Kaveh Hadiloo, Parsa Mostanadi, Ali Asadzadeh, Siavash Taremi, Abdolreza Esmaeilzadeh
{"title":"Targeting cancer stem cells with CAR-based immunotherapy: biology, evidence, and future directions.","authors":"Kaveh Hadiloo, Parsa Mostanadi, Ali Asadzadeh, Siavash Taremi, Abdolreza Esmaeilzadeh","doi":"10.1186/s12935-025-03846-3","DOIUrl":"10.1186/s12935-025-03846-3","url":null,"abstract":"<p><p>Cancer stem cells (CSCs) are pivotal in tumor initiation, progression, and relapse, underscoring the need for targeted therapies to achieve lasting responses. This review delves into CSC biology, highlighting their tumor-initiating potential demonstrated through limiting dilution assays and their role in resistance to therapies. Although successful CAR therapies, such as anti-CD19 CAR T-cells, can induce complete responses without directly targeting CSCs, CAR strategies focusing on CSCs may offer promising avenues to prevent recurrence. We assess CAR therapies targeting CSC-specific antigens, including CD133 and GD2, in preclinical and clinical contexts, emphasizing their effectiveness against glioblastoma, breast cancer, and other malignancies. Nevertheless, challenges such as marker specificity and suppression by the tumor microenvironment (TME) persist. Future strategies, which may include dual-targeting and AI-driven marker discovery, aim to improve CSC elimination and advance personalized cancer immunotherapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"289"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12302805/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728097","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
Targeting m6A methylation for early diagnosis and precision medicine in hepatocellular carcinoma. 靶向m6A甲基化用于肝细胞癌的早期诊断和精准医疗。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03923-7
Qing-Kang Zheng, Ya-Nan Shi, Ming-Yuan Yang, Yi-Yuan Xie, Kai Sun, Huan-Zhang Niu
{"title":"Targeting m6A methylation for early diagnosis and precision medicine in hepatocellular carcinoma.","authors":"Qing-Kang Zheng, Ya-Nan Shi, Ming-Yuan Yang, Yi-Yuan Xie, Kai Sun, Huan-Zhang Niu","doi":"10.1186/s12935-025-03923-7","DOIUrl":"10.1186/s12935-025-03923-7","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) ranks as a significant global malignancy, occupying the sixth position in incidence and the third in cancer-related mortality. Despite this, the mechanisms underlying HCC progression remain insufficiently understood. m6A modification is one of the most common post-transcriptional modifications in eukaryotic mRNA, regulated by methyltransferases, demethylases, and m<sup>6</sup>A-binding proteins. Proteins specialized in m6A recognition selectively bind to m6A-modified RNA, influencing processes such as splicing, maturation, nucleation, degradation, and translation. Current research, both domestic and international, primarily explores how m6A modification and its associated proteins affect malignant cell proliferation, migration, invasion, metastasis, and drug resistance. However, the role of m6A-related proteins in tumor progression in HCC remains poorly characterized. This review elucidates the composition, mechanisms, and biological functions of m6A methylation modification proteins in HCC progression, alongside recent advancements in m6A-related biomarker discovery and immunotherapeutic developments, aiming to enhance early clinical diagnosis and facilitate targeted drug development for HCC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"286"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12302885/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728099","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
m6A-modified CTC-297N7.9 inhibits hepatocellular carcinoma metastasis via epigenetically downregulating CCL2 and CD47. m6a修饰的CTC-297N7.9通过表观遗传下调CCL2和CD47抑制肝癌转移。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03857-0
Huamei Wei, Changhong Pu, Min Zeng, Rongzhou Lu, Yunyu Wei, Yanyan Huang, Zheng Huang, Lizheng Huang, Zuoming Xu, Jianchu Wang, Rihai Ma, Jian Pu
{"title":"m<sup>6</sup>A-modified CTC-297N7.9 inhibits hepatocellular carcinoma metastasis via epigenetically downregulating CCL2 and CD47.","authors":"Huamei Wei, Changhong Pu, Min Zeng, Rongzhou Lu, Yunyu Wei, Yanyan Huang, Zheng Huang, Lizheng Huang, Zuoming Xu, Jianchu Wang, Rihai Ma, Jian Pu","doi":"10.1186/s12935-025-03857-0","DOIUrl":"10.1186/s12935-025-03857-0","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is one of most common malignancies with poor prognosis, largely due to its high propensity for metastasis and recurrence. As the most common internal RNA modification, N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) plays critical and diverse roles in HCC progression. However, the mechanisms by which m<sup>6</sup>A regulates HCC metastasis remain incompletely understood. Here, we aimed to identify key m<sup>6</sup>A modification events during HCC metastasis.</p><p><strong>Methods: </strong>The expression of CTC-297N7.9. was measured by qPCR. m<sup>6</sup>A modification level of CTC-297N7.9 was measured by methylated RNA immunoprecipitation (MeRIP) and single-base elongation- and ligation-based qPCR amplification method (SELECT). The roles of m<sup>6</sup>A-modified CTC-297N7.9 in HCC were investigated by in vitro cell viability, proliferation, migration and phagocytosis assays, and in vivo liver metastasis and lung metastasis assays. The underlying mechanisms of m<sup>6</sup>A-modified CTC-297N7.9 were dissected by chromatin isolation by RNA purification (ChIRP), assay for transposase accessible chromatin (ATAC) and cleavage under target & tagmentation (CUT&Tag) assays.</p><p><strong>Results: </strong>The m<sup>6</sup>A modification level of CTC-297N7.9 is decreased in HCC tissues and correlated with microvascular invasion and poor prognosis. CTC-297N7.9 suppresses HCC metastasis in an m<sup>6</sup>A-dependent manner. m<sup>6</sup>A-modified CTC-297N7.9 attenuates tumor-associated macrophages (TAMs) infiltration and M2 polarization through downregulating CCL2 expression and secretion. Additionally, m<sup>6</sup>A-modified CTC-297N7.9 promotes phagocytosis of HCC cells by macrophages through downregulating the phagocytosis checkpoint CD47. Mechanistic investigations revealed that m<sup>6</sup>A-modified CTC-297N7.9 binds the m<sup>6</sup>A reader YTHDC1, which further binds and recruits the histone H3K9me3 methyltransferase SETDB1 and H3K27me3 methyltransferase EZH2 to the promoters of CCL2 and CD47, leading to the upregulation of H3K9me3 and H3K27me3 modifications at CCL2 and CD47 promoters, and transcriptional silencing of CCL2 and CD47.</p><p><strong>Conclusions: </strong>Our study demonstrates that m<sup>6</sup>A-modified CTC-297N7.9 acts as a metastasis suppressor in HCC, and highlights its potential as a prognostic biomarker and therapeutic target for HCC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"290"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12306055/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728095","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
Identification of CD4_Naive cells related gene FMO4 as a positive regulator of the poor prognosis of septic CRC patients. CD4_Naive细胞相关基因FMO4作为脓毒性结直肠癌患者不良预后的正调节因子的鉴定
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03917-5
Weiye Hou, Peiwen Fu, Zhenling Nie, Wanye Wang, Jingjing Li, Bangshun He, Qiao Tang, Tianyi Gao
{"title":"Identification of CD4_Naive cells related gene FMO4 as a positive regulator of the poor prognosis of septic CRC patients.","authors":"Weiye Hou, Peiwen Fu, Zhenling Nie, Wanye Wang, Jingjing Li, Bangshun He, Qiao Tang, Tianyi Gao","doi":"10.1186/s12935-025-03917-5","DOIUrl":"10.1186/s12935-025-03917-5","url":null,"abstract":"<p><strong>Background: </strong>Septic disease usually results in delayed access to intensive care and poor outcomes in CRC patients. Studies have shown that T cells play important roles in CRC and sepsis immune systems. Hence, this study was performed to investigate the role of T cells and T-cell-related genes in CRC-related sepsis prognosis.</p><p><strong>Methods: </strong>Single-cell sequencing data from CRC and sepsis patients were first analysed to explore common T-cell signals, including those related to cellular communication and signalling pathways. Then, functional enrichment analysis and Mendelian randomization (MR) analysis were conducted on marker genes of T cells to identify the key genes and their effects on septic CRC. The expression of key genes and their associations with CRC prognosis were validated in 32 blood samples from CRC patients with sepsis.</p><p><strong>Results: </strong>Compared with that in the negative control group, CD4_Naive cells infiltration was significantly lower in the combined single-cell sequencing data analysis of CRC and sepsis patients (p < 0.05). Our MR analysis and clinical sample verification results revealed that a high FMO4 gene was negatively associated with CD4_Naive cells infiltration and related to poor prognosis in septic patients with CRC (p < 0.05). The functional enrichment analysis of FMO4 revealed that FMO4 participated mainly in xenobiotic catabolic processes and taurine and hypo-Taurine metabolism in septic CRC, which further inhibited the energy metabolic activity of CD4_Naive cells.</p><p><strong>Conclusion: </strong>The CD4_Naive cells related gene FMO4 appears to promote poor prognosis in septic CRC patients, suggesting that it is a promising candidate for therapeutic intervention.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"285"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12302463/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728093","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
IGFBP1 orchestrates metabolic reprogramming to drive clear cell renal cell carcinoma progression through NR1H4-mediated lipid homeostasis disruption. IGFBP1协调代谢重编程,通过nr1h4介导的脂质稳态破坏驱动透明细胞肾细胞癌的进展。
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03904-w
Yanning Sun, Yuhu Hao, Fan Peng, Hongju Ling, Kai Sun, Jiechuan Qiu, Tianmin Yang, Leizuo Zhao, Qinghua Xia
{"title":"IGFBP1 orchestrates metabolic reprogramming to drive clear cell renal cell carcinoma progression through NR1H4-mediated lipid homeostasis disruption.","authors":"Yanning Sun, Yuhu Hao, Fan Peng, Hongju Ling, Kai Sun, Jiechuan Qiu, Tianmin Yang, Leizuo Zhao, Qinghua Xia","doi":"10.1186/s12935-025-03904-w","DOIUrl":"10.1186/s12935-025-03904-w","url":null,"abstract":"<p><p>The hallmark lipid accumulation phenotype observed in clear cell renal cell carcinoma (ccRCC) serves as a critical pathophysiological driver of tumor progression. Our investigation revealed that IGFBP1 expression was significantly elevated in ccRCC versus matched normal renal tissues, with increased levels correlating with poorer patient survival outcomes. IGFBP1 knockdown not only suppressed tumor proliferation and invasiveness in vitro but also provoked substantial lipidomic remodeling, as validated through comprehensive lipidomic profiling. Specifically, IGFBP1-deficient cells demonstrated marked reductions in triglycerides (TGs), diacylglycerols (DAGs), free fatty acids (FFA), and cholesterol esters (CEs), thereby establishing IGFBP1 as a key regulator of the metabolic derangements' characteristic of ccRCC pathogenesis. Mechanistic exploration identified NR1H4 as a potential transcriptional regulator operating downstream of IGFBP1-mediated signaling pathways. A thorough interrogation of these pathways established mechanistic links between IGFBP1 activity and endoplasmic reticulum stress, revealing an integrated network that coordinates lipid homeostasis within malignant renal epithelium. These findings substantiated the role of IGFBP1 as a central node in the metabolic reprogramming associated with ccRCC and propose actionable targets for therapeutic intervention through modulation of lipid metabolic pathways.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"284"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12302755/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728094","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
Construction and validation of acetylation-related gene signatures for immune landscape analysis and prognostication risk prediction in luminal breast cancer. 用于腔内乳腺癌免疫景观分析和预后风险预测的乙酰化相关基因标记的构建和验证
IF 6 2区 医学
Cancer Cell International Pub Date : 2025-07-28 DOI: 10.1186/s12935-025-03920-w
Mengdi Zhu, Jinna Lin, Haohan Liu, Jingru Wang, Nianqiu Liu, Yudong Li, Hongna Lai, Qianfeng Shi
{"title":"Construction and validation of acetylation-related gene signatures for immune landscape analysis and prognostication risk prediction in luminal breast cancer.","authors":"Mengdi Zhu, Jinna Lin, Haohan Liu, Jingru Wang, Nianqiu Liu, Yudong Li, Hongna Lai, Qianfeng Shi","doi":"10.1186/s12935-025-03920-w","DOIUrl":"10.1186/s12935-025-03920-w","url":null,"abstract":"<p><strong>Background: </strong>Epigenetic acetylation plays an essential role in the development and drug resistance of luminal breast cancer. However, the acetylation regulatory network in luminal breast cancer remains underexplored.</p><p><strong>Methods: </strong>We used the TCGA-BRCA database to explore the acetylation regulatory network in luminal breast cancer. Spearman correlation coefficients, Cox proportional hazards, and the STRING database were used to identify genes that were correlated with acetylation regulatory molecules in luminal breast cancer and could predict patient outcomes. An acetylation regulatory risk model was constructed via Consensus Cluster Plus and the LASSO risk model. GSEA, K‒M survival analysis, and receiver operating characteristic (ROC) curve analysis were used to analyze survival and possible regulatory pathways of the risk model. TIDE, Microenvironment Cell Populations-counter, and CIBERSORT algorithms were used to analyze the immune landscape of the risk model population. Patients' tumor specimens were used to detect the expression of KAT2B and TAF1L. The luminal breast cancer cell lines MCF-7 and T47D were used in cell viability, Transwell, western blotting, and RT‒qPCR experiments to confirm the risk model. Mouse model was constructed for in vivo validation of KAT2B and TAF1L function.</p><p><strong>Results: </strong>In our study, we utilized the TCGA-BRCA database to conduct a comprehensive analysis of the acetylation regulatory pattern in luminal breast cancer. Using Consensus Cluster Plus and the LASSO risk model, we screened 6 acetylation-related genes (KAT2B, TAF1L, CDC37, CCDC107, C17orf106, and ASPSCR1) and constructed a 6-gene risk model of luminal breast cancer. Based on this model, luminal breast cancer patients were classified into high- and low-risk subgroups. The high-risk subgroup had a poor prognosis. Further analysis revealed that the high-risk subgroup was associated with lower CD8 + T-cell infiltration and greater responsiveness to immune checkpoint inhibitor therapy. In vitro and in vivo experiments revealed that knockdown of KAT2B and TAF1L dramatically inhibited tumor cell proliferation. In vitro experiments also showed knockdown of KAT2B and TAF1L dramatically inhibited tumor cell migration, increased lymphocyte infiltration, and significantly upregulated the expression of CD8 + T-cell-associated chemokines in luminal breast cancer cells.</p><p><strong>Conclusions: </strong>In this study, we successfully constructed a 6-gene acetylation-associated risk model for luminal breast cancer, providing a new direction and evidence for personalized treatment. Our results also suggested that KAT2B and TAF1L might serve as potential therapeutic targets in luminal breast cancer.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"25 1","pages":"287"},"PeriodicalIF":6.0,"publicationDate":"2025-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12302894/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144728092","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