1
|
Huang G, Chen S, Han B, Zhang G, Bao M, Paka Lubamba G, Hua Y, Li H, Liu W, Shen J, Wang L, Lin J, Tang PMK, Ding Z, Li C. Apolipoprotein D is crucial for promoting perineural invasion in salivary adenoid cystic carcinoma. Br J Cancer 2025; 132:599-610. [PMID: 39962257 PMCID: PMC11962118 DOI: 10.1038/s41416-025-02946-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/22/2024] [Accepted: 01/16/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Perineural invasion (PNI) is a prevalent phenomenon in salivary adenoid cystic carcinoma (SACC). Nevertheless, the regulatory mechanism of PNI is largely elusive. METHODS We detected Apolipoprotein D (ApoD) expression and further determined its role in SACC progression. Subsequently, the contributions of SACC-derived ApoD on neurite outgrowth of dorsal root ganglions (DRGs) cells were explored. Moreover, a series of in vivo assays were conducted to elucidate the role of ApoD in the SACC PNI process. RESULTS We observed a dramatic up-regulation of ApoD in the SACC associated with an enhancement of PNI in patient biopsies. We found that SACC-derived ApoD elevated cancer cell migration and invasion. In addition, ApoD could facilitate the neurite outgrowth of cultured DRG cells in a CXCR4-dependent manner in vitro, as well as innervation, angiogenesis, and invasion along peripheral nerves of SACC in vivo. More importantly, by advanced bioinformatic analysis, we unexpectedly revealed a novel phenomenon 'tumour cell to neuron-like cell transition' in the ApoD-rich microenvironment in vivo, contributing to the neurogenesis in the SACC tumour. CONCLUSION we discovered a novel role of cancer-derived ApoD in the pathogenesis of PNI, which may represent an effective therapeutic target for SACC in clinics.
Collapse
Affiliation(s)
- Guangzhao Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Su Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Bo Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Gaowei Zhang
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Mingzhe Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Grace Paka Lubamba
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yufei Hua
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Honglin Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Wenwen Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, 100081, Beijing, China
| | - Jiefei Shen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Lei Wang
- Department of Dentistry, The Second People's Hospital of Tibet Autonomous Region, Xizang, 850030, China
| | - Jie Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Anesthesiology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhangfan Ding
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Chunjie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Deng MH, Yang XW, Zhou YM, Xie LZ, Zou T, Ping JG. In silico research of coagulation- and fibrinolysis-related genes for predicting prognosis of clear cell renal cell carcinoma. Transl Androl Urol 2025; 14:307-324. [PMID: 40114841 PMCID: PMC11921444 DOI: 10.21037/tau-24-483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Background Coagulation- and fibrinolysis-related genes (CFRGs) are involved in tumor progression. However, their regulatory mechanisms in clear cell renal cell carcinoma (ccRCC) remain unclear. The aim of this study was to search for genes related to coagulation and fibrinolytic systems in ccRCC and to investigate their potential role in tumor pathogenesis and progression. Methods Differentially expressed genes (DEGs) between ccRCC and control samples, as well as key module genes associated with ccRCC, were extracted from The Cancer Genome Atlas-Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) dataset. Differentially expressed CFRGs (DE-CFRGs) were identified by intersecting these DEGs with CFRGs. Prognostic genes were identified through univariate Cox, least absolute shrinkage and selection operator (LASSO), and multivariate Cox analyses of DE-CFRGs. Additional independent prognostic and enrichment analyses were conducted, and potential therapeutic drugs were predicted. In addition, quantitative real-time polymerase chain reaction (RT-qPCR) was performed to validate the expression of prognostic genes. Results Sixteen DE-CFRGs were identified by intersecting 3,311 DEGs, 1,719 key module genes, and CFRGs. Four prognostic genes-TIMP1, RUNX1, BMP6, and PROS1-were found to be involved in complement and coagulation cascades and other functional pathways. The prognostic model demonstrated strong predictive power for ccRCC, with stage, risk score, and grade all correlating with prognosis. Additionally, 14 potential drugs, such as tamoxifen citrate and cytarabine, were predicted for therapeutic targeting of the identified prognostic genes. RT-qPCR confirmed that the expression levels of TIMP1, and RUNX1 were significantly upregulated in ccRCC samples, consistent with bioinformatics analysis. Conclusions A prognostic model incorporating TIMP1, RUNX1, BMP6, and PROS1 was constructed, offering new insights for prognostic evaluation and therapeutic strategies in ccRCC.
Collapse
Affiliation(s)
- Ming-Hao Deng
- Department of Urology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Xue-Wen Yang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu-Ming Zhou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Lv-Zhong Xie
- Department of Urology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Tao Zou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ji-Gen Ping
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Peng Z, Fang W, Wu B, He M, Li S, Wei J, Hao Y, Jin L, Liu M, Zhang X, Wei Y, Ge Y, Wei Y, Qian H, Zhang Y, Jiang J, Chang Z, Rao Y, Zhang X, Cui CP, Zhang L. Targeting Smurf1 to block PDK1-Akt signaling in KRAS-mutated colorectal cancer. Nat Chem Biol 2025; 21:59-70. [PMID: 39039255 DOI: 10.1038/s41589-024-01683-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 06/23/2024] [Indexed: 07/24/2024]
Abstract
The phosphoinositide 3-kinase (PI3K)-Akt axis is one of the most frequently activated pathways and is demonstrated as a therapeutic target in Kirsten rat sarcoma viral oncogene homolog (KRAS)-mutated colorectal cancer (CRC). Targeting the PI3K-Akt pathway has been a challenging undertaking through the decades. Here we unveiled an essential role of E3 ligase SMAD ubiquitylation regulatory factor 1 (Smurf1)-mediated phosphoinositide-dependent protein kinase 1 (PDK1) neddylation in PI3K-Akt signaling and tumorigenesis. Upon growth factor stimulation, Smurf1 immediately triggers PDK1 neddylation and the poly-neural precursor cell expressed developmentally downregulated protein 8 (poly-Nedd8) chains recruit methyltransferase SET domain bifurcated histone lysine methyltransferase 1 (SETDB1). The cytoplasmic complex of PDK1 assembled with Smurf1 and SETDB1 (cCOMPASS) consisting of PDK1, Smurf1 and SETDB1 directs Akt membrane attachment and T308 phosphorylation. Smurf1 deficiency dramatically reduces CRC tumorigenesis in a genetic mouse model. Furthermore, we developed a highly selective Smurf1 degrader, Smurf1-antagonizing repressor of tumor 1, which exhibits efficient PDK1-Akt blockade and potent tumor suppression alone or combined with PDK1 inhibitor in KRAS-mutated CRC. The findings presented here unveil previously unrecognized roles of PDK1 neddylation and offer a potential strategy for targeting the PI3K-Akt pathway and KRAS mutant cancer therapy.
Collapse
Affiliation(s)
- Zhiqiang Peng
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Shanghai Fengxian Central Hospital, The Third School of Clinical Medicine, Southern Medical University, Shanghai, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Wei Fang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Shanghai Fengxian Central Hospital, The Third School of Clinical Medicine, Southern Medical University, Shanghai, China
| | - Bo Wu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Ming He
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China
| | - Shaohua Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Shanghai Fengxian Central Hospital, The Third School of Clinical Medicine, Southern Medical University, Shanghai, China
| | - Jun Wei
- Shanghai Fengxian Central Hospital, The Third School of Clinical Medicine, Southern Medical University, Shanghai, China
| | - Yang Hao
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Lujia Jin
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Mingqiu Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xin Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yange Wei
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yingwei Ge
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yinghua Wei
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yangjun Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Junyi Jiang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhijie Chang
- School of Medicine, Tsinghua University, Beijing, China
| | - Yu Rao
- MOE Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, China.
| | - Xueli Zhang
- Shanghai Fengxian Central Hospital, The Third School of Clinical Medicine, Southern Medical University, Shanghai, China.
| | - Chun-Ping Cui
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| |
Collapse
|
4
|
Wang H, Liu C, Jin K, Li X, Zheng J, Wang D. Research advances in signaling pathways related to the malignant progression of HSIL to invasive cervical cancer: A review. Biomed Pharmacother 2024; 180:117483. [PMID: 39353319 DOI: 10.1016/j.biopha.2024.117483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The progression of high-grade squamous intraepithelial lesion (HSIL) to invasive cervical cancer (ICC) is a complex process involving persistent human papillomavirus (HPV) infection and changes in signal transduction regulation, energy and material metabolism, cell proliferation, autoimmune, and other biological process in vaginal microenvironment and immune microenviroment. Signaling pathways are a series of interacting molecules in cells that regulate various physiological functions of cells, such as growth, differentiation, metabolism, and death. In the progression of HSIL to ICC, abnormal activation or inhibition in signaling pathways plays an essensial role. This review presented some signaling pathways related to the malignant progression of HSIL to ICC, including p53, Rb, PI3K/AKT/mTOR, Wnt/β-catenin, Notch, NF-κB, MAPK, TGF-β, JAK-STAT, Hippo, and Hedgehog. The molecular mechanisms involved in the biological process of pathway regulation were also analyzed, in order to illustrate the molecular pathway of HSIL progression to ICC and provide references for the development of more effective prevention and treatment methods.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Obstetrics and Gynecology, Quanzhou Medical College, Quanzhou, Fujian 362010, China
| | - Chang Liu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Keer Jin
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Xiang Li
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Jiaxin Zheng
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Danbo Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| |
Collapse
|
5
|
Xu J, Yang W, Xie X, Gu C, Zhao L, Liu F, Zhang N, Bai Y, Liu D, Liu H, Jin X, Meng Y. Identification of 10 differentially expressed genes involved in the tumorigenesis of cervical cancer via next-generation sequencing. PeerJ 2024; 12:e18157. [PMID: 39372720 PMCID: PMC11453159 DOI: 10.7717/peerj.18157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
Background The incidence and mortality of cervical cancer remain high in female malignant tumors worldwide. There is still a lack of diagnostic and prognostic markers for cervical carcinoma. This study aimed to screen differentially expressed genes (DEGs) between normal and cervical cancer tissues to identify candidate genes for further research. Methods Uterine cervical specimens were resected from our clinical patients after radical hysterectomy. Three patients' transcriptomic datasets were built by the next generation sequencing (NGS) results. DEGs were selected through the edgeR and DESeq2 packages in the R environment. Functional enrichment analysis, including GO/DisGeNET/KEGG/Reactome enrichment analysis, was performed. Normal and cervical cancer tissue data from the public databases TCGA and GTEx were collected to compare the expression levels of 10 selected DEGs in tumor and normal tissues. ROC curve and survival analysis were performed to compare the diagnostic and prognostic values of each gene. The expression levels of candidate genes were verified in 15 paired clinical specimens via quantitative real-time polymerase chain reaction. Results There were 875 up-regulated and 1,482 down-regulated genes in cervical cancer samples compared with the paired adjacent normal cervical tissues according to the NGS analysis. The top 10 DEGs included APOD, MASP1, ACKR1, C1QTNF7, SFRP4, HSPB6, GSTM5, IGFBP6, F10 and DCN. GO, DisGeNET and Reactome analyses revealed that the DEGs were related to extracellular matrix and angiogenesis which might influence tumorigenesis. KEGG enrichment showed that PI3K-Akt signaling pathway might be involved in cervical cancer tumorigenesis and progression. The expression levels of selected genes were decreased in tumors in both the public database and our experimental clinical specimens. All the candidate genes showed excellent diagnostic value, and the AUC values exceeded 0.90. Additionally, APOD, ACKR1 and SFRP4 expression levels could help predict the prognosis of patients with cervical cancer. Conclusions In this study, we selected the top 10 DEGs which were down-regulated in cervical cancer tissues. All of them had dramatically diagnostic value. APOD, ACKR1 and SFRP4 were associated with the survivals of cervical cancer. C1QTNF7, HSPB6, GSTM5, IGFBP6 and F10 were first reported to be candidate genes of cervical carcinoma.
Collapse
Affiliation(s)
- Jia Xu
- School of Medicine, Nankai University, Tianjin, China
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wen Yang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiufeng Xie
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chenglei Gu
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Luyang Zhao
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Feng Liu
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Nina Zhang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuge Bai
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Dan Liu
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hainan Liu
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiangshu Jin
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuanguang Meng
- School of Medicine, Nankai University, Tianjin, China
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Jiang R, Li X, Cao S, Wu Y, Zhang W, Huang Y. Nodal staging score for adequacy of nodal staging in cervical cancer. Heliyon 2024; 10:e26116. [PMID: 38596019 PMCID: PMC11001770 DOI: 10.1016/j.heliyon.2024.e26116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/03/2024] [Accepted: 02/07/2024] [Indexed: 04/11/2024] Open
Abstract
Background Cervical cancer remains the fourth most common female malignancy with increasing newly cases around the world. It is of clinical value to precisely evaluate whether false negative nodal existed and develop a nodal staging model in cervical cancer. Materials and methods Clinical data of cervical cancer patients was retrieved from the Surveillance, Epidemiology, and End Results database. Probability of missing nodal disease and nodal staging score (NSS) was computed to assess the nodal status of each individual.Prognostic value of NSS was assessed. Results A total of 9056 individuals were in this study, with 5115 squamous cell carcinoma, 2791 adenocarcinoma, 512 adenosquamous carcinoma, and 638 other type individuals. A beta-binomial model was used to compute the probability of nodal disease in four histological types, respectively. False negative probability drastically decreased as more nodes examined. To reach 0.05 of false negative probability, it required at least 17 lymph nodes in squamous cell carcinoma patients,18 in adenocarcinoma, 12 in adenosquamous carcinoma patients and 14 in other types. To reach 0.95 of NSS, it took 10 lymph nodes in squamous cell carcinoma, 6 in adenocarcinoma, 10 in adenosquamous carcinoma and 7 in other types. Significant prognostic values of NSS quartiles subsets were found in all four histological sets. Conclusion NSS tool enables adequate nodal staging of cervical cancer with significant prognostic value. Exact number of lymph nodes required for surgery in cervical cancer is specified based on histologic type.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaoqi Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Siyu Cao
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yong Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Wei Zhang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yan Huang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| |
Collapse
|
7
|
Song C, Liu M, Wang G, Xu Z, Han S, Peng M, Liu L, Wang W, Xu Y, Ni Q, He J. BMP6 inhibits gastric cancer growth and predicts good prognosis. J Gastrointest Oncol 2024; 15:22-32. [PMID: 38482225 PMCID: PMC10932659 DOI: 10.21037/jgo-23-512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 12/03/2023] [Indexed: 02/19/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common tumors in the digestive tract, and effective treatment methods are still lacking. Bone morphogenetic protein 6 (BMP6) is closely related to the occurrence and development of various tumors, but its relevance to GC is still unclear. The aim of the study was to explore the relationship between BMP6 and the occurrence and development of GC. METHODS In this study, we investigated the relationship between BMP6 and the prognosis of GC patients using bioinformatics technology and clinical tissue samples. We also explored the connection between BMP6 and the biological behavior of GC cells through molecular biology experiments and relevant in vivo animal experiments. Finally, we examined the mechanisms by which BMP6 inhibits the onset and progression of GC. RESULTS Through analysis of The Cancer Genomics Atlas (TCGA) database, we observed that BMP6 is expressed at low levels in GC, and its low expression is associated with a poor prognosis in GC patients. Cell experiments demonstrated that BMP6 expression can influence the proliferation of GC cells both in vitro and in vivo. Furthermore, we discovered that BMP6 is linked to the nuclear factor-κB (NF-κB) pathway, and subsequent experiments confirmed that BMP6 can inhibit the biological activity of GC cells by activating the NF-κB pathway. CONCLUSIONS Our findings suggest that BMP6 is a potential prognostic biomarker in GC and can regulate the biological activity of GC cells through the NF-κB pathway. BMP6 may serve as a promising therapeutic target for GC, and our study introduces novel ideas for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Chao Song
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, Shanghai, China
| | - Mengmeng Liu
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, Shanghai, China
| | - Ganggang Wang
- Department of General Surgery, Pudong Hospital, Fudan University, Shanghai, China
| | - Zijin Xu
- Department of General Surgery, Affiliated Zhongshan Hospital of Fudan University, Qingpu Branch, Shanghai, China
| | - Siyang Han
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Maozhen Peng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianzhi Ni
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Junyi He
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Liu S, Yao Y, Hou M, Mei J, Sun L, Zhang G. Identification and validation of a ferroptosis-related signature for prediction of the prognosis and tumor microenvironment in patients with chromophobe renal cell carcinoma. BMC Cancer 2023; 23:1079. [PMID: 37940859 PMCID: PMC10634106 DOI: 10.1186/s12885-023-11589-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Ferroptosis is a novel form of regulated cell death that is different from other forms, which has an important role in tumor growth inhibition. The purpose of this study was to construct and validate a prognostic signature related to ferroptosis in chromophobe renal cell carcinoma (ChRCC) and to explore its role in immune cell infiltration and systemic therapy. METHODS The gene expression profiles of ChRCC patients obtained from The Cancer Genome Atlas (TCGA) database were used to identify differentially expressed prognostic ferroptosis-related genes (FRGs) by univariate Cox proportional hazards analyses. Ferroptosis molecular subtypes were obtained by consensus clustering analysis. The FRG-based signature in the training set was established by least absolute shrinkage and selection operator analysis and verified in the testing set. The association between molecular subtypes and the prognostic signature and immune microenvironment was explored to predict responses to immunotherapy. Immunohistochemistry was used to verify expression of the FRG-based signature externally. RESULTS ChRCC patients were divided into two FRG subtypes. Two FRGs (TFRC and SLC7A11) were identified to construct the prognostic signature. The high-risk group and cluster 2 had worse overall survival than the low-risk group and cluster 1, respectively. The low-risk group and cluster 1 had higher levels of immune cell infiltration and expression of MHC and immune checkpoint molecules than the high-risk group and cluster 2. The risk score was a predictor of overall survival and had a good predictive ability, which was verified in the testing set and evaluated by ROC and calibration curves. The high-risk group had a higher tumor mutation burden. The different sensitivities of targeted drugs in patients with different risks were evaluated. External immunohistochemical analysis showed that TFRC and SLC7A11 were highly expressed in tumor tissues compared with para-cancer normal tissues, and the expression level was significantly associated with a more advanced stage and worse cancer-specific survival. CONCLUSIONS An FRG signature was identified and validated to predict the clinicopathological features and prognosis of ChRCC. A significant association between the signature and immune cell infiltration, immune checkpoint expression, and drug response is helpful to guide comprehensive treatment of ChRCC.
Collapse
Affiliation(s)
- Shuai Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Rd, 266003, Qingdao, P.R. China
| | - Yu Yao
- Department of Urology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Rd, 266003, Qingdao, P.R. China
| | - Mingyu Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, 266003, Qingdao, P.R. China
| | - Jingchang Mei
- Department of Urology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Rd, 266003, Qingdao, P.R. China
| | - Lijiang Sun
- Department of Urology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Rd, 266003, Qingdao, P.R. China
| | - Guiming Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Rd, 266003, Qingdao, P.R. China.
| |
Collapse
|
9
|
Han S, Liu X, Ju S, Mu W, Abulikemu G, Zhen Q, Yang J, Zhang J, Li Y, Liu H, Chen Q, Cui B, Wu S, Zhang Y. New mechanisms and biomarkers of lymph node metastasis in cervical cancer: reflections from plasma proteomics. Clin Proteomics 2023; 20:35. [PMID: 37689639 PMCID: PMC10492398 DOI: 10.1186/s12014-023-09427-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 08/21/2023] [Indexed: 09/11/2023] Open
Abstract
OBJECTIVE Lymph node metastasis (LNM) and lymphatic vasculature space infiltration (LVSI) in cervical cancer patients indicate a poor prognosis, but satisfactory methods for diagnosing these phenotypes are lacking. This study aimed to find new effective plasma biomarkers of LNM and LVSI as well as possible mechanisms underlying LNM and LVSI through data-independent acquisition (DIA) proteome sequencing. METHODS A total of 20 cervical cancer plasma samples, including 7 LNM-/LVSI-(NC), 4 LNM-/LVSI + (LVSI) and 9 LNM + /LVSI + (LNM) samples from a cohort, were subjected to DIA to identify differentially expressed proteins (DEPs) for LVSI and LNM. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed for DEP functional annotation. Protein-protein interaction (PPI) and weighted gene coexpression network analysis (WGCNA) were used to detect new effective plasma biomarkers and possible mechanisms. RESULTS A total of 79 DEPs were identified in the cohort. GO and KEGG analyses showed that DEPs were mainly enriched in the complement and coagulation pathway, lipid and atherosclerosis pathway, HIF-1 signal transduction pathway and phagosome and autophagy. WGCNA showed that the enrichment of the green module differed greatly between groups. Six interesting core DEPs (SPARC, HPX, VCAM1, TFRC, ERN1 and APMAP) were confirmed to be potential plasma diagnostic markers for LVSI and LNM in cervical cancer patients. CONCLUSION Proteomic signatures developed in this study reflected the potential plasma diagnostic markers and new possible pathogenesis mechanisms in the LVSI and LNM of cervical cancer.
Collapse
Affiliation(s)
- Sai Han
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaoli Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Shuang Ju
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Wendi Mu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Gulijinaiti Abulikemu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Qianwei Zhen
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Jiaqi Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Jingjing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Yi Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Hongli Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Baoxia Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China
| | - Shuxia Wu
- Department of Obstetrics and Gynecology, the Fifth People's Hospital of Jinan, Jinan, Shandong, 250012, People's Republic of China.
| | - Youzhong Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
10
|
Chen Y, Wang Y, Li Z, Jing J, Jiang D, Yuan X, Li F. Exploration of the Mechanism of Shengxian Decoction Against Chronic Obstructive Pulmonary Disease Based on Network Pharmacology and Experimental Verification. Assay Drug Dev Technol 2023; 21:258-272. [PMID: 37682969 DOI: 10.1089/adt.2023.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023] Open
Abstract
Shengxian decoction (SXT) is clinically used in chronic obstructive pulmonary disease (COPD) treatment. This study aimed to explore the mechanism and target genes of SXT acting on COPD. Differentially expressed genes (DEGs) between COPD and controls were identified and then performed enrichment analysis. The effective active compounds and corresponding target genes were obtained from the traditional Chinese medicine systems pharmacology database. We also compiled COPD related genes from the GeneCards database. Through the protein-protein interaction (PPI) network and least absolute shrinkage and selection operator (LASSO) regression was performed to identify key genes. Molecular docking was used for docking of key genes and compounds. The expression of key genes was detected by quantitative real-time PCR in COPD patients and bronchial epithelial cells stimulated with cigarette stroke extract (CSE). We identified 1,458 intersected DEGs from GSE47460 and GSE57148 datasets. Compared with intersected DEGs, we obtained 33 SXT target COPD-related genes. PI3K-Akt signaling pathway, MAPK signaling pathway, and focal adhesion were enriched by these 33 genes, as well as intersected DEGs. According to LASSO regression, there were 12 genes considered as signature genes. Then we constructed active compounds and corresponding six target genes. Finally, HIF1A and IL1B were selected as key genes by combining PPI network. HIF1A and IL1B were all upregulated expression in COPD and CSE stimulated cells and recovered in SXT treated CSE stimulated cells. This study provides a scientific basis for the identification of active compounds and target genes of SXT in the treatment of COPD.
Collapse
Affiliation(s)
- Yifei Chen
- Basic Teaching and Research Office of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Shuimogou, Urumqi, China
| | - Yiming Wang
- Department of Acupuncture, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
| | - Zheng Li
- Department of Respiration, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
- Department of Respiration, National Clinical Research Base of Traditional Chinese Medicine in Xinjiang, Shaybagh, Urumqi, China
| | - Jing Jing
- Department of Respiration, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
- Department of Respiration, National Clinical Research Base of Traditional Chinese Medicine in Xinjiang, Shaybagh, Urumqi, China
| | - De Jiang
- Basic Teaching and Research Office of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Shuimogou, Urumqi, China
| | - Xiaoxia Yuan
- Basic Teaching and Research Office of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Xinjiang Medical University, Shuimogou, Urumqi, China
| | - Fengsen Li
- Department of Respiration, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Shaybagh, Urumqi, China
| |
Collapse
|
11
|
Zhang C, Gu L, Xiao J, Jin F. Knockdown of RBM15 inhibits tumor progression and the JAK-STAT signaling pathway in cervical cancer. BMC Cancer 2023; 23:684. [PMID: 37474926 PMCID: PMC10360283 DOI: 10.1186/s12885-023-11163-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/08/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND RNA binding motif protein 15 (RBM15), a writer of N6-methyladenosine (m6A) methylation, contributes significantly to the development of various tumors. However, the function of RBM15 in cervical cancer (CC) has not been determined. METHODS Based on the GSE9750, GSE63514, and m6A datasets, m6A-related differentially expressed genes (DEGs) were screened out. The hub genes were identified by generating a Protein-Protein Interaction (PPI) network. RT-qPCR was conducted to assess the mRNA expression of hub genes. CCK8, scratch wound healing, and transwell assays were utilized to examine the influence of RBM15 on HeLa and SiHa cells. Tumor xenograft models were used to assess the effects of RBM15 on tumorigenesis. A mechanistic analysis of RBM15 in CC tumors was conducted using the GeneCards and Coxpresdb databases, followed by a Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and the pathway-related genes were subsequently validated using Western blotting. RESULTS Five DEGs were screened, including WTAP, RBM15, CBLL1, and YTHDC2. Among them, WTAP, RBM15, CBLL1, and YTHDC2 were hub genes and can be used as biomarkers for CC. RBM15 expression was considerably increased, while WTAP, CBLL1, and YTHDC2 were significantly downregulated. Knockdown of RBM15 significantly suppressed the proliferation, invasion, and migration of CC cells and tumorigenesis. Moreover, knockdown of RBM15 significantly reduced the expression levels of proteins related to the JAK-STAT pathway. CONCLUSIONS Knockdown of RBM15 inhibited the progression of CC cells, which probably by inhibiting the JAK-STAT pathway pathway.
Collapse
Affiliation(s)
- Chunnian Zhang
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China.
| | - Liqin Gu
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China
| | - Juan Xiao
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China
| | - Feng Jin
- Department of Gynecology, Ganzhou People's Hospital, No. 16, Meiguan Avenue, Ganzhou City, 341000, Jiangxi Province, China
| |
Collapse
|
12
|
Wu M, Wang Z, Zhang J. Analysis on tumor immune microenvironment and construction of a prognosis model for immune-related skin cutaneous melanoma. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:671-681. [PMID: 37539569 PMCID: PMC10930407 DOI: 10.11817/j.issn.1672-7347.2023.230069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVES Malignant melanoma is a highly malignant and heterogeneous skin cancer. Although immunotherapy has improved survival rates, the inhibitory effect of tumor microenvironment has weakened its efficacy. To improve survival and treatment strategies, we need to develop immune-related prognostic models. Based on the analysis of the Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and Sequence Read Archive (SRA) database, this study aims to establish an immune-related prognosis prediction model, and to evaluate the tumor immune microenvironment by risk score to guide immunotherapy. METHODS Skin cutaneous melanoma (SKCM) transcriptome sequencing data and corresponding clinical information were obtained from the TCGA database, differentially expressed genes were analyzed, and prognostic models were developed using univariate Cox regression, the LASSO method, and stepwise regression. Differentially expressed genes in prognostic models confirmed by real-time reverse transcription PCR (real-time RT-PCR) and Western blotting. Survival analysis was performed by using the Kaplan-Meier method, and the effect of the model was evaluated by time-dependent receiver operating characteristic curve as well as multivariate Cox regression, and the prognostic model was validated by 2 GEO melanoma datasets. Furthermore, correlations between risk score and immune cell infiltration, Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) score, immune checkpoint mRNA expression levels, tumor immune cycle, or tumor immune micro-environmental pathways were analyzed. Finally, we performed association analysis for risk score and the efficacy of immunotherapy. RESULTS We identified 4 genes that were differentially expressed in TCGA-SKCM datasets, which were mainly associated with the tumor immune microenvironment. A prognostic model was also established based on 4 genes. Among 4 genes, the mRNA and protein levels of killer cell lectin like receptor D1 (KLRD1), leukemia inhibitory factor (LIF), and cellular retinoic acid binding protein 2 (CRABP2) genes in melanoma tissues differed significantly from those in normal skin (all P<0.01). The prognostic model was a good predictor of prognosis for patients with SKCM. The patients with high-risk scores had significantly shorter overall survival than those with low-risk scores, and consistent results were achieved in the training cohort and multiple validation cohorts (P<0.001). The risk score was strongly associated with immune cell infiltration, ESTIMATE score, immune checkpoint mRNA expression levels, tumor immune cycle, and tumor immune microenvironmental pathways (P<0.001). The correlation analysis showed that patients with the high-risk scores were in an inhibitory immune microenvironment based on the prognostic model (P<0.01). CONCLUSIONS The immune-related SKCM prognostic model constructed in this study can effectively predict the prognosis of SKCM patients. Considering its close correlation to the tumor immune microenvironment, the model has some reference value for clinical immunotherapy of SKCM.
Collapse
Affiliation(s)
- Meng Wu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008.
- Department of Dermatology, Hunan Provincial People's Hospital, Changsha 410002.
| | - Zheng Wang
- School of Computer Science, Hunan First Normal University, Changsha 410205. w8614@ hotmail.com
| | - Jianglin Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008.
- Department of Dermatology, Shenzhen People's Hospital, Shenzhen Guangdong 518020, China.
| |
Collapse
|
13
|
Chen C, Cao Y, Li W, Liu Z, Liu P, Tian X, Sun C, Wang W, Gao H, Kang S, Wang S, Jiang J, Chen C, Tian J. The pathological risk score: A new deep learning-based signature for predicting survival in cervical cancer. Cancer Med 2022; 12:1051-1063. [PMID: 35762423 PMCID: PMC9883425 DOI: 10.1002/cam4.4953] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 05/26/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To develop and validate a deep learning-based pathological risk score (RS) with an aim of predicting patients' prognosis to investigate the potential association between the information within the whole slide image (WSI) and cervical cancer prognosis. METHODS A total of 251 patients with the International Federation of Gynecology and Obstetrics (FIGO) Stage IA1-IIA2 cervical cancer who underwent surgery without any preoperative treatment were enrolled in this study. Both the clinical characteristics and WSI of each patient were collected. To construct a prognosis-associate RS, high-dimensional pathological features were extracted using a convolutional neural network with an autoencoder. With the score threshold selected by X-tile, Kaplan-Meier survival analysis was applied to verify the prediction performance of RS in overall survival (OS) and disease-free survival (DFS) in both the training and testing datasets, as well as different clinical subgroups. RESULTS For the OS and DFS prediction in the testing cohort, RS showed a Harrell's concordance index of higher than 0.700, while the areas under the curve (AUC) achieved up to 0.800 in the same cohort. Furthermore, Kaplan-Meier survival analysis demonstrated that RS was a potential prognostic factor, even in different datasets or subgroups. It could further distinguish the survival differences after clinicopathological risk stratification. CONCLUSION In the present study, we developed an effective signature in cervical cancer for prognosis prediction and patients' stratification in OS and DFS.
Collapse
Affiliation(s)
- Chi Chen
- Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine, School of Medicine and EngineeringBeihang UniversityBeijingChina,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex SystemsInstitute of Automation, Chinese Academy of SciencesBeijingChina
| | - Yuye Cao
- Department of Obstetrics and GynecologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Weili Li
- Department of Obstetrics and GynecologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex SystemsInstitute of Automation, Chinese Academy of SciencesBeijingChina,School of Artificial IntelligenceUniversity of Chinese Academy of SciencesBeijingChina
| | - Ping Liu
- Department of Obstetrics and GynecologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xin Tian
- Department of Obstetrics and GynecologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Caixia Sun
- Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine, School of Medicine and EngineeringBeihang UniversityBeijingChina,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex SystemsInstitute of Automation, Chinese Academy of SciencesBeijingChina
| | - Wuliang Wang
- Department of Obstetrics and GynecologyThe Second Affiliated Hospital of He' nan Medical UniversityZhengzhouChina
| | - Han Gao
- Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine, School of Medicine and EngineeringBeihang UniversityBeijingChina,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex SystemsInstitute of Automation, Chinese Academy of SciencesBeijingChina
| | - Shan Kang
- Department of GynecologyFourth Hospital Hebei Medical UniversityShijiazhuangChina
| | - Shaoguang Wang
- Department of GynecologyYantai Yuhuangding HospitalYantaiChina
| | - Jingying Jiang
- Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine, School of Medicine and EngineeringBeihang UniversityBeijingChina,Key Laboratory of Big Data‐Based Precision Medicine (Beihang University)Ministry of Industry and Information TechnologyBeijingChina
| | - Chunlin Chen
- Department of Obstetrics and GynecologyNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jie Tian
- Beijing Advanced Innovation Center for Big Data‐Based Precision Medicine, School of Medicine and EngineeringBeihang UniversityBeijingChina,CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex SystemsInstitute of Automation, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
14
|
Zhong QH, Zha SW, Lau ATY, Xu YM. Recent knowledge of NFATc4 in oncogenesis and cancer prognosis. Cancer Cell Int 2022; 22:212. [PMID: 35698138 PMCID: PMC9190084 DOI: 10.1186/s12935-022-02619-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Nuclear factor of activated T-cells, cytoplasmic 4 (NFATc4), a transcription factor of NFAT family, which is activated by Ca2+/calcineurin signaling. Recently, it is reported that aberrantly activated NFATc4 participated and modulated in the initiation, proliferation, invasion, and metastasis of various cancers (including cancers of the lung, breast, ovary, cervix, skin, liver, pancreas, as well as glioma, primary myelofibrosis and acute myelocytic leukemia). In this review, we cover the latest knowledge on NFATc4 expression pattern, post-translational modification, epigenetic regulation, transcriptional activity regulation and its downstream targets. Furthermore, we perform database analysis to reveal the prognostic value of NFATc4 in various cancers and discuss the current unexplored areas of NFATc4 research. All in all, the result from these studies strongly suggest that NFATc4 has the potential as a molecular therapeutic target in multiple human cancer types.
Collapse
Affiliation(s)
- Qiu-Hua Zhong
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041 People’s Republic of China
| | - Si-Wei Zha
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041 People’s Republic of China
| | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041 People’s Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041 People’s Republic of China
| |
Collapse
|
15
|
Lian L, Teng SB, Xia YY, Shen XM, Zheng Y, Han SG, Wang WJ, Xu XF, Zhou C. Development and verification of a hypoxia- and immune-associated prognosis signature for esophageal squamous cell carcinoma. J Gastrointest Oncol 2022; 13:462-477. [PMID: 35557566 PMCID: PMC9086047 DOI: 10.21037/jgo-22-69] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/08/2022] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Esophageal cancer is one of the most common gastrointestinal malignancies worldwide, with high morbidity and mortality in China. The clinical importance of the interaction between hypoxia and immune status in the tumor microenvironment has been established in esophageal squamous cell carcinoma (ESCC). This study aims to develop a new hypoxia- and immune-based gene signature to predict the survival of ESCC patients. METHODS The RNA-sequencing and clinical data of 173 cases of ESCC and 271 normal tissues were obtained from The Cancer Genome Atlas (TCGA) data portal and the Genotype-Tissue Expression (GTEx) database. Hypoxia-related genes (HRGs) and immune-related genes (IRGs) were retrieved from publicly shared data. Differentially expressed gene (DEG) analyses were carried out by the DESeq2 method using the edgeR package in R. Based on the intersection of the DEGs and HRGs/IRGs, differentially expressed HRGs (DEHRGs) and differentially expressed IRGs (DEIRGs) were obtained. DEHRGs and DEIRGs associated with prognosis were evaluated using univariate Cox proportional hazards analysis. A prognostic risk score model was constructed according to the genes acquired through Cox regression. Univariate analysis and Cox proportional hazards analysis were used to determine the independent prognostic factors related to prognosis. A nomogram was developed to predict the 1-, 2-, and 3-year overall survival (OS) probability. RESULTS A total of 73 intersecting genes were obtained as DEHRGs and a total of 548 intersecting genes were obtained as DEIRGs. The risk score was established using 8 genes (FABP7, TLR1, SYTL1, APLN, OSM, EGFR, IL17RD, MYH9) acquired from univariate Cox analysis. Based on this 8-gene-based risk score, a risk prognosis classifier was constructed to classify the samples into high- and low-risk groups according to the median risk score. The nomogram model was constructed to predict the OS of ESCC patients. CONCLUSIONS The hypoxia- and immune-based gene signature might serve as a prognostic classifier for clinical decision-making regarding individualized management, follow-up plans, and treatment strategies for ESCC patients.
Collapse
Affiliation(s)
- Lian Lian
- Department of Oncology, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Shi-Bing Teng
- Department of Thoracic Surgery, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - You-You Xia
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang, China
| | - Xiao-Ming Shen
- Department of Oncology, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Yan Zheng
- Department of Oncology, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Shu-Guang Han
- Department of General Surgery, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Wen-Jie Wang
- Department of Radio-Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Xue-Fei Xu
- Department of General Surgery, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Chong Zhou
- Department of Radiation Oncology, Xuzhou Central Hospital, Xuzhou, China
| |
Collapse
|
16
|
Regulation of stability and inhibitory activity of the tumor suppressor SEF through casein-kinase II-mediated phosphorylation. Cell Signal 2021; 86:110085. [PMID: 34280495 DOI: 10.1016/j.cellsig.2021.110085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/23/2022]
Abstract
Inflammation and cancer are intimately linked. A key mediator of inflammation is the transcription-factor NF-κB/RelA:p50. SEF (also known as IL-17RD) is a feedback antagonist of NF-κB/RelA:p50 that is emerging as an important link between inflammation and cancer. SEF acts as a buffer to prevent excessive NF-κB activity by sequestering NF-κB/RelA:p50 in the cytoplasm of unstimulated cells, and consequently attenuating the NF-κB response upon pro-inflammatory cytokine stimulation. SEF contributes to cancer progression also via modulating other signaling pathways, including those triggered by growth-factors. Despite its important role in human physiology and pathology, mechanisms that regulate SEF biochemical properties and inhibitory activity are unknown. Here we show that human SEF is an intrinsically labile protein that is stabilized via CK2-mediated phosphorylation, and identified the residues whom phosphorylation by CK2 stabilizes hSEF. Unlike endogenous SEF, ectopic SEF was rapidly degraded when overexpressed but was stabilized in the presence of excess CK2, suggesting a mechanism for limiting SEF levels depending upon CK2 processivity. Additionally, phosphorylation by CK2 potentiated hSef interaction with NF-κB in cell-free binding assays. Most importantly, we identified a CK2 phosphorylation site that was indispensable for SEF inhibition of pro-inflammatory cytokine signaling but was not required for SEF inhibition of growth-factor signaling. To our knowledge, this is the first demonstration of post-translational modifications that regulate SEF at multiple levels to optimize its inhibitory activity in a specific signaling context. These findings may facilitate the design of SEF variants for treating cytokine-dependent pathologies, including cancer and chronic inflammation.
Collapse
|
17
|
Li L, Gao H, Wang D, Jiang H, Wang H, Yu J, Jiang X, Huang C. Metabolism-Relevant Molecular Classification Identifies Tumor Immune Microenvironment Characterization and Immunotherapeutic Effect in Cervical Cancer. Front Mol Biosci 2021; 8:624951. [PMID: 34277697 PMCID: PMC8280349 DOI: 10.3389/fmolb.2021.624951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 06/14/2021] [Indexed: 12/19/2022] Open
Abstract
Cervical cancer (CESC) is a gynecologic malignant tumor associated with high incidence and mortality rates because of its distinctive management complexity. Herein, we characterized the molecular features of CESC based on the metabolic gene expression profile by establishing a novel classification system and a scoring system termed as METAscore. Integrative analysis was performed on human CESC samples from TCGA dataset. Unsupervised clustering of RNA sequencing data on 2,752 formerly described metabolic genes identified three METAclusters. These METAclusters for overall survival time, immune characteristics, metabolic features, transcriptome features, and immunotherapeutic effectiveness existed distinct differences. Then we analyzed 207 DEGs among the three METAclusters and as well identified three geneclusters. Correspondingly, these three geneclusters also differently expressed among the aforementioned features, supporting the reliability of the metabolism-relevant molecular classification. Finally METAscore was constructed which emerged as an independent prognostic biomarker, related to CESC transcriptome features, metabolic features, immune characteristics, and linked to the sensitivity of immunotherapy for individual patient. These findings depicted a new classification and a scoring system in CESC based on the metabolic pattern, thereby furthering the understanding of CESC genetic signatures and aiding in the prediction of the effectiveness to anticancer immunotherapies.
Collapse
Affiliation(s)
- Luyi Li
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China.,The 2 Afflicated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui Gao
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Danhan Wang
- The 2 Afflicated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hao Jiang
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| | - Hongzhu Wang
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| | - Jiajian Yu
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| | - Xin Jiang
- Prenatal Diagnosis Center of NanFang Hospital, The Southern Medical University, Guangzhou, China
| | - Changjiang Huang
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|