1
|
Tong Z, Wang Z, Jiang J, Fu W, Hu S. Glycyrrhizin enhances the antitumor activity of cisplatin in non‑small cell lung cancer cells by influencing DNA damage and apoptosis. Oncol Lett 2025; 29:207. [PMID: 40070780 PMCID: PMC11894513 DOI: 10.3892/ol.2025.14954] [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: 09/18/2024] [Accepted: 01/07/2025] [Indexed: 03/14/2025] Open
Abstract
The objective of the present study was to elucidate the mechanism by which glycyrrhizin enhances the antitumor activity of cisplatin in non-small cell lung cancer. Initially, A549 cells were treated with different concentrations of glycyrrhizin (0.25-8 mM) or cisplatin (10-160 µM) for 48 h to investigate the effect of glycyrrhizin combined with cisplatin on A549 cells in vitro. Subsequently, A549 cells were divided into control (untreated), CP (20 µM cisplatin), GL (2 mM glycyrrhizin) and CP + GL (20 µM cisplatin + 2 mM glycyrrhizin) groups to elucidate the underlying mechanism of glycyrrhizin. After 48 h incubation, the viability and colony-forming ability of the cells were assessed using MTT and colony formation assays. Apoptosis levels and cell cycle progression were analyzed using flow cytometry and western blotting was used to evaluate apoptosis- and cell cycle-related proteins. Additionally, comet assays and western blotting were used to evaluate DNA damage and relevant proteins. The results demonstrated both glycyrrhizin and cisplatin individually reduced A549 cell viability in a concentration-dependent manner. Cisplatin demonstrated a lower half-maximal inhibitory concentration (IC50) at higher glycyrrhizin concentrations, with an IC50 value of ~35 µM with 2 mM glycyrrhizin. Furthermore, the combined treatment of glycyrrhizin and cisplatin synergistically reduced cell colony-forming ability, induced apoptosis and arrested the cell cycle at the G2 phase, showing greater efficacy when compared with either treatment individually. In addition, western blotting analysis demonstrated that, in comparison with treatment with cisplatin or glycyrrhizin alone, the combined treatment markedly increased the protein expression levels of B-cell lymphoma 2-associated X protein, cleaved-caspase-3/caspase-3, γH2AX, phosphorylated-checkpoint kinase 1 and phosphorylated-p53/p53, while notably reducing the protein levels of B-cell lymphoma 2, cyclin D1, cyclin-dependent kinase 2 and cyclin-dependent kinase 4. The findings of the present study indicate that glycyrrhizin enhances the antitumor efficacy of cisplatin in non-small cell lung cancer cells by modulating DNA damage and apoptosis.
Collapse
Affiliation(s)
- Zhufeng Tong
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Zhen Wang
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Jinghan Jiang
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Wenqi Fu
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Siying Hu
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| |
Collapse
|
2
|
Han Q, Gu Y, Xiang H, Zhang L, Wang Y, Yang C, Li J, Steiner C, Lapalombella R, Woyach JA, Yang Y, Dovat S, Song C, Ge Z. Targeting WDR5/ATAD2 signaling by the CK2/IKAROS axis demonstrates therapeutic efficacy in T-ALL. Blood 2025; 145:1407-1421. [PMID: 39785511 PMCID: PMC11969266 DOI: 10.1182/blood.2024024130] [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: 02/13/2024] [Revised: 10/21/2024] [Accepted: 11/06/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy with a poor prognosis and limited options for targeted therapies. Identifying new molecular targets to develop novel therapeutic strategies is the pressing immediate issue in T-ALL. Here, we observed high expression of WD repeat-containing protein 5 (WDR5) in T-ALL. With in vitro and in vivo models, we demonstrated the oncogenic role of WDR5 in T-ALL by activating cell cycle signaling through its new downstream effector, ATPase family AAA domain-containing 2 (ATAD2). Moreover, the function of a zinc finger transcription factor of the Kruppel family (IKAROS) is often impaired by genetic alteration and casein kinase II (CK2) which is overexpressed in T-ALL. We found that IKAROS directly regulates WDR5 transcription; CK2 inhibitor, CX-4945, strongly suppresses WDR5 expression by restoring IKAROS function. Last, combining CX-4945 with WDR5 inhibitor demonstrates synergistic efficacy in the patient-derived xenograft mouse models. In conclusion, our results demonstrated that WDR5/ATAD2 is a new oncogenic signaling pathway in T-ALL, and simultaneous targeting of WRD5 and CK2/IKAROS has synergistic antileukemic efficacy and represents a promising potential strategy for T-ALL therapy.
Collapse
Affiliation(s)
- Qi Han
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Yan Gu
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Huimin Xiang
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Linyao Zhang
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Yan Wang
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Chan Yang
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Jun Li
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| | - Chelsea Steiner
- Division of Hematology, The Ohio State University Wexner Medical Center, The James Cancer Hospital, Columbus, OH
| | - Rosa Lapalombella
- Division of Hematology, The Ohio State University Wexner Medical Center, The James Cancer Hospital, Columbus, OH
| | - Jennifer A. Woyach
- Division of Hematology, The Ohio State University Wexner Medical Center, The James Cancer Hospital, Columbus, OH
| | - Yiping Yang
- Division of Hematology, The Ohio State University Wexner Medical Center, The James Cancer Hospital, Columbus, OH
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Hershey Medical Center, Pennsylvania State University Medical College, Hershey, PA
| | - Chunhua Song
- Division of Hematology, The Ohio State University Wexner Medical Center, The James Cancer Hospital, Columbus, OH
| | - Zheng Ge
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing, China
| |
Collapse
|
3
|
Fan T, Xiao C, Deng Z, Li S, Tian H, Zheng Y, Zheng B, Li C, He J. Signatures of H3K4me3 modification predict cancer immunotherapy response and identify a new immune checkpoint-SLAMF9. Respir Res 2025; 26:17. [PMID: 39815269 PMCID: PMC11734478 DOI: 10.1186/s12931-024-03093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025] Open
Abstract
H3 lysine 4 trimethylation (H3K4me3) modification and related regulators extensively regulate various crucial transcriptional courses in health and disease. However, the regulatory relationship between H3K4me3 modification and anti-tumor immunity has not been fully elucidated. We identified 72 independent prognostic genes of lung adenocarcinoma (LUAD) whose transcriptional expression were closely correlated with known 27 H3K4me3 regulators. We constructed three H3K4me3 modification patterns utilizing the expression profiles of the 72 genes, and patients classified in each pattern exhibited unique tumor immune infiltration characteristics. Using the principal component analysis (PCA) of H3K4me3-related patterns, we constructed a H3K4me3 risk score (H3K4me3-RS) system. The deep learning analysis using 12,159 cancer samples from 26 cancer types and 725 cancer samples from 5 immunotherapy cohorts revealed that H3K4me3-RS was significantly correlated with cancer immune tolerance and sensitivity. Importantly, this risk-score system showed satisfactory predictive performance for the ICB therapy responses of patients suffering from several cancer types, and we identified that SLAMF9 was one of the immunosuppressive phenotype and immunotherapy resistance-determined genes of H3K4me3-RS. The mice melanoma model showed Slamf9 knockdown remarkably restrained cancer progression and enhanced the efficacy of anti-CTLA-4 and anti-PD-L1 therapies by elevating CD8 + T cell infiltration. This study provided a new H3K4me3-associated biomarker system to predict tumor immunotherapy response and suggested the preclinical rationale for investigating the roles of SLAMF9 in cancer immunity regulation and treatment.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuofeng Li
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
4
|
Li X, You Q. Sanguinarine identified as a natural dual inhibitor of AURKA and CDK2 through network pharmacology and bioinformatics approaches. Sci Rep 2024; 14:29608. [PMID: 39609491 PMCID: PMC11605095 DOI: 10.1038/s41598-024-81063-0] [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: 08/02/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
Cervical cancer (CA) continues to be a female malignant tumor with limited therapeutic options, resulting in a high mortality rate. Sanguinarine (SANG), a naturally occurring alkaloid, has demonstrated notable efficacy in preclinical treatment of CA. However, the mechanism through which SANG acts against CA is not fully understood. To address this, utilizing nine drug target prediction databases, we have successfully identified 379 potential targets for SANG. Venn diagram analysis compared 2367 CA-related targets from the GeneCards disease database, 2618 CA-closely related targets derived from multiple datasets in GEO through WGCNA analysis, and the 379 potential targets of SANG, resulting in 35 shared targets. Subsequently, by employing PPI network analysis, the Cytohubba plugin, the Human Protein Atlas, TCGA database data, and ROC curve analysis, we have identified AURKA and CDK2 as key targets of SANG in combating CA. Single-gene GSEA results suggest that the overexpression of AURKA and CDK2 is closely correlated with DNA replication, cell cycle progression, and various DNA repair pathways in CA. Molecular docking and molecular simulation dynamics analyses have confirmed the stable binding of both AURKA and CDK2 to SANG. In summary, by integrating diverse methodological approaches, this study discovered that SANG potentially inhibits the malignant features of CA by targeting AURKA and CDK2, thereby regulating DNA replication, cell cycle progression, and multiple DNA repair pathways. This lays a solid foundation for further exploring the pharmacological role of SANG in CA therapy. However, further in-depth in vitro and in vivo experiments are required to corroborate our findings.
Collapse
Affiliation(s)
- Xiang Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Qi You
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
5
|
Cheng L, Yang C, Lu J, Huang M, Xie R, Lynch S, Elfman J, Huang Y, Liu S, Chen S, He B, Lin T, Li H, Chen X, Huang J. Oncogenic SLC2A11-MIF fusion protein interacts with polypyrimidine tract binding protein 1 to facilitate bladder cancer proliferation and metastasis by regulating mRNA stability. MedComm (Beijing) 2024; 5:e685. [PMID: 39156764 PMCID: PMC11324686 DOI: 10.1002/mco2.685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/03/2024] [Accepted: 07/14/2024] [Indexed: 08/20/2024] Open
Abstract
Chimeric RNAs, distinct from DNA gene fusions, have emerged as promising therapeutic targets with diverse functions in cancer treatment. However, the functional significance and therapeutic potential of most chimeric RNAs remain unclear. Here we identify a novel fusion transcript of solute carrier family 2-member 11 (SLC2A11) and macrophage migration inhibitory factor (MIF). In this study, we investigated the upregulation of SLC2A11-MIF in The Cancer Genome Atlas cohort and a cohort of patients from Sun Yat-Sen Memorial Hospital. Subsequently, functional investigations demonstrated that SLC2A11-MIF enhanced the proliferation, antiapoptotic effects, and metastasis of bladder cancer cells in vitro and in vivo. Mechanistically, the fusion protein encoded by SLC2A11-MIF interacted with polypyrimidine tract binding protein 1 (PTBP1) and regulated the mRNA half-lives of Polo Like Kinase 1, Roundabout guidance receptor 1, and phosphoinositide-3-kinase regulatory subunit 3 in BCa cells. Moreover, PTBP1 knockdown abolished the enhanced impact of SLC2A11-MIF on biological function and mRNA stability. Furthermore, the expression of SLC2A11-MIF mRNA is regulated by CCCTC-binding factor and stabilized through RNA N4-acetylcytidine modification facilitated by N-acetyltransferase 10. Overall, our findings revealed a significant fusion protein orchestrated by the SLC2A11-MIF-PTBP1 axis that governs mRNA stability during the multistep progression of bladder cancer.
Collapse
Affiliation(s)
- Liang Cheng
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Chenwei Yang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Junlin Lu
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Ming Huang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Clinical Research Center for Urological DiseasesDepartment of Urology, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Ruihui Xie
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Clinical Research Center for Urological DiseasesDepartment of Urology, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Sarah Lynch
- Department of PathologySchool of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Justin Elfman
- Department of PathologySchool of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Yuhang Huang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Sen Liu
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Siting Chen
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Baoqing He
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Tianxin Lin
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Clinical Research Center for Urological DiseasesDepartment of Urology, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hui Li
- Department of PathologySchool of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Xu Chen
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Clinical Research Center for Urological DiseasesDepartment of Urology, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Jian Huang
- Department of UrologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationDepartment of Urology,Sun Yat‐sen Memorial Hospital,Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Guangdong Provincial Clinical Research Center for Urological DiseasesDepartment of Urology, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
6
|
Zhang S, Lin T, Xiong X, Chen C, Tan P, Wei Q. Targeting histone modifiers in bladder cancer therapy - preclinical and clinical evidence. Nat Rev Urol 2024; 21:495-511. [PMID: 38374198 DOI: 10.1038/s41585-024-00857-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2024] [Indexed: 02/21/2024]
Abstract
Bladder cancer in the most advanced, muscle-invasive stage is lethal, and very limited therapeutic advances have been reported for decades. To date, cisplatin-based chemotherapy remains the first-line therapy for advanced bladder cancer. Late-line options have historically been limited. In the past few years, next-generation sequencing technology has enabled chromatin remodelling gene mutations to be characterized, showing that these alterations are more frequent in urothelial bladder carcinoma than in other cancer types. Histone modifiers have functional roles in tumour progression by modulating the expression of tumour suppressors and oncogenes and, therefore, have been considered as novel drug targets for cancer therapy. The roles of epigenetic reprogramming through histone modifications have been increasingly studied in bladder cancer, and the therapeutic efficacy of targeting those histone modifiers genetically or chemically is being assessed in preclinical studies. Results from preclinical studies in bladder cancer encouraged the investigation of some of these drugs in clinical trials, which yield mixed results. Further understanding of how alterations of histone modification mechanistically contribute to bladder cancer progression, drug resistance and tumour microenvironment remodelling will be required to facilitate clinical application of epigenetic drugs in bladder cancer.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tianhai Lin
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xingyu Xiong
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chong Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Ping Tan
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Wang J, Tan Z, Huang Y, Li C, Zhan P, Wang H, Li H. Integrating single-cell RNA-seq to identify fibroblast-based molecular subtypes for predicting prognosis and therapeutic response in bladder cancer. Aging (Albany NY) 2024; 16:11385-11408. [PMID: 39033778 PMCID: PMC11315389 DOI: 10.18632/aging.206021] [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: 01/09/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Bladder cancer (BLCA) is a highly aggressive and heterogeneous disease, posing challenges for diagnosis and treatment. Cancer immunotherapy has recently emerged as a promising option for patients with advanced and drug-resistant cancers. Fibroblasts, a significant component of the tumor microenvironment, play a crucial role in tumor progression, but their precise function in BLCA remains uncertain. METHODS Single-cell RNA sequencing (scRNA-seq) data for BLCA were obtained from the Gene Expression Omnibus database. The R package "Seurat" was used for processing scRNA-seq data, with uniform manifold approximation and projection (UMAP) for downscaling and cluster identification. The FindAllMarkers function identified marker genes for each cluster. Differentially expressed genes influencing overall survival (OS) of BLCA patients were identified using the limma package. Differences in clinicopathological characteristics, immune microenvironment, immune checkpoints, and chemotherapeutic drug sensitivity between high- and low-risk groups were investigated. RT-qPCR and immunohistochemistry validated the expression of prognostic genes. RESULTS Fibroblast marker genes identified three molecular subtypes in the testing set. A prognostic signature comprising ten genes stratified BLCA patients into high- and low-score groups. This signature was validated in one internal and two external validation sets. High-score patients exhibited increased immune cell infiltration, elevated chemokine expression, and enhanced immune checkpoint expression but had poorer OS and a reduced response to immunotherapy. Six sensitive anti-tumor drugs were identified for the high-score group. RT-qPCR and immunohistochemistry showed that CERCAM, TM4SF1, FN1, ANXA1, and LOX were highly expressed, while EMP1, HEYL, FBN1, and SLC2A3 were downregulated in BLCA. CONCLUSION A novel fibroblast marker gene-based signature was established, providing robust predictions of survival and immunotherapeutic response in BLCA patients.
Collapse
Affiliation(s)
- Jia Wang
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Endocrinology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhiyong Tan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yinglong Huang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Charles Li
- Core Facility for Protein Research, Chinese Academy of Sciences, Beijing, China
- Zhongke Jianlan Medical Research Institute, Beijing, China
- Zhejiang Institute of Integrated Traditional and Western Medicine, Hangzhou, China
| | - Peiqin Zhan
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haifeng Wang
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haihao Li
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
8
|
Wu J, Huang M, Dong W, Chen Y, Zhou Q, Zhang Q, Zheng J, Liu Y, Zhang Y, Liu S, Yang C, Chen S, Huang J, Lin T, Chen X. SUMO E3 ligase MUL1 inhibits lymph node metastasis of bladder cancer by mediating mitochondrial HSPA9 translocation. Int J Biol Sci 2024; 20:3986-4006. [PMID: 39113711 PMCID: PMC11302872 DOI: 10.7150/ijbs.98772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Lymph node (LN) metastasis is the dominant cause of death in bladder cancer (BCa) patients, but the underlying mechanism remains largely unknown. In recent years, accumulating studies have confirmed that bidirectional mitochondria-nucleus communication is essential for sustaining multiple function of mitochondria. However, little has been studied regarding whether and how the translocation of mitochondrial proteins is involved in LN metastasis. In this study, we first identified that the SUMO E3 ligase MUL1 was significantly downregulated in LN-metastatic BCa tissues and correlated with a good prognosis. Mechanistically, MUL1 SUMOylated HSPA9 at the K612 residue, leading to HSPA9 export from mitochondria and interaction with SUZ12 and in the nucleus. Consequently, MUL1 induced the ubiquitination-mediated degradation of SUZ12 and EZH2 and induced downstream STAT3 pathway inhibition in a HSPA9-dependent manner. Importantly, mutation of HSPA9 SUMO-conjugation motifs limited the translocation of mitochondrial HSPA9 and blocked the HSPA9-SUZ12 and HSPA9-EZH2 interactions. With mutation of the HSPA9 K612 site, the suppressive role of MUL1 overexpression was lost in BCa cells. Further in vitro and in vivo assays revealed that MUL1 inhibits the metastasis and proliferation of BCa cells. Overall, our study reveals a novel function and molecular mechanism of SUMO E3 ligases in LN metastasis.
Collapse
Affiliation(s)
- Jilin Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Ming Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Wen Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Yuelong Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Qiang Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Junjiong Zheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Yeqing Liu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Yangjie Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Sen Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Chenwei Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Siting Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
9
|
Qing L, Li Q, Yang Y, Xu W, Wang Y, Li R, You C, Dong Z. Hypoxia-mediated attenuation of EGLN2 inhibition of the NF-κB signaling pathway leads to the formation of a loop between HIF-1α and MUC1-C promoting chemoresistance in bladder cancer. Mol Carcinog 2024; 63:1303-1318. [PMID: 38634741 DOI: 10.1002/mc.23725] [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: 10/17/2023] [Revised: 01/30/2024] [Accepted: 03/29/2024] [Indexed: 04/19/2024]
Abstract
The expression pattern of MUC1-C in tumors is closely linked to tumor progression; however, its specific mechanism remains unclear. The expression of MUC1-C in cancer and adjacent normal tissues was detected using immunohistochemistry and Western blot. The IC50 of cells to gemcitabine was determined using the CCK8 assay. The effects of hypoxia and MUC1-C on the behavioral and metabolic characteristics of bladder cancer cells were investigated. Gene expression was assessed through Western blot and polymerase chain reaction. The relationship between the genes was analyzed by co-immunoprecipitation, immunofluorescence and Western blot. Finally, the role of the EGLN2 and NF-κB signaling pathways in the interaction between MUC1-C and hypoxia-inducible factor-1α (HIF-1α) was investigated. MUC1-C expression is significantly higher in bladder cancer tissues than in adjacent normal tissues, particularly in large-volume tumors, and is closely correlated with clinical features such as tumor grade. Tumor volume-mediated hypoxia resulted in increased expression of MUC1-C and HIF-1α in bladder cancer cells. Under stimulation of hypoxia, the inhibitory effect of EGLN2 on the NF-κB signaling pathway was weakened, allowing NF-κB to promote the positive feedback formation of MUC1-C and HIF-1α. Simultaneously, EGLN2-mediated degradation of HIF-1α was reduced. This ultimately led to elevated HIF-1α-mediated downstream gene expression, promoting increased glucose uptake and glycolysis, and ultimately resulting in heightened chemotherapy resistance and malignancy.
Collapse
Affiliation(s)
- Liangliang Qing
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Qingchao Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yongjin Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenbo Xu
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yanan Wang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Rongxing Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Chengyu You
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhilong Dong
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
- Key Laboratory of Urological Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
10
|
Tan Y, Qiao J, Yang S, Liu H, Wang Q, Liu Q, Feng W, Cui L. H3K4me3-Mediated FOXJ2/SLAMF8 Axis Aggravates Thrombosis and Inflammation in β2GPI/Anti-β2GPI-Treated Monocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309140. [PMID: 38639399 PMCID: PMC11199983 DOI: 10.1002/advs.202309140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/31/2024] [Indexed: 04/20/2024]
Abstract
Antiphospholipid syndrome (APS) is characterized by thrombus formation, poor pregnancy outcomes, and a proinflammatory response. H3K4me3-related monocytes activation are key regulators of APS pathogenesis. Therefore, H3K4me3 CUT&Tag and ATAC-seq are performed to examine the epigenetic profiles. The results indicate that the H3K4me3 signal and chromatin accessibility at the FOXJ2 promoter are enhanced in an in vitro monocyte model by stimulation with β2GPI/anti-β2GPI, which mimics APS, and decreases after OICR-9429 administration. Furthermore, FOXJ2 is highly expressed in patients with primary APS (PAPS) and is the highest in patients with triple-positive antiphospholipid antibodies (aPLs). Mechanistically, FOXJ2 directly binds to the SLAMF8 promoter and activates SLAMF8 transcription. SLAMF8 further interacts with TREM1 to stimulate TLR4/NF-κB signaling and prohibit autophagy. Knockdown of FOXJ2, SLAMF8, or TREM1 blocks TLR4/NF-κB and provokes autophagy, subsequently inhibiting the release of inflammatory and thrombotic indicators. A mouse model of vascular APS is established via β2GPI intraperitoneal injection, and the results suggest that OICR-9429 administration attenuates the inflammatory response and thrombus formation by inactivating FOXJ2/SLAMF8/TREM1 signaling. These findings highlight the overexpression of H3K4me3-mediated FOXJ2 in APS, which consequently accelerates APS pathogenesis by triggering inflammation and thrombosis via boosting the SLAMF8/TREM1 axis. Therefore, OICR-9429 is a promising candidate drug for APS therapy.
Collapse
Affiliation(s)
- Yuan Tan
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Jiao Qiao
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Shuo Yang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Hongchao Liu
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qingchen Wang
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Qi Liu
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Weimin Feng
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| | - Liyan Cui
- Institute of Medical TechnologyPeking University Health Science CenterBeijing100191China
- Department of Laboratory MedicinePeking University Third HospitalBeijing100191China
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijing100191China
| |
Collapse
|
11
|
Hofvander J, Qiu A, Lee K, Bilenky M, Carles A, Cao Q, Moksa M, Steif J, Su E, Sotiriou A, Goytain A, Hill LA, Singer S, Andrulis IL, Wunder JS, Mertens F, Banito A, Jones KB, Underhill TM, Nielsen TO, Hirst M. Synovial Sarcoma Chromatin Dynamics Reveal a Continuum in SS18:SSX Reprograming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594262. [PMID: 38798672 PMCID: PMC11118320 DOI: 10.1101/2024.05.14.594262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Synovial sarcoma (SyS) is an aggressive soft-tissue malignancy characterized by a pathognomonic chromosomal translocation leading to the formation of the SS18::SSX fusion oncoprotein. SS18::SSX associates with mammalian BAF complexes suggesting deregulation of chromatin architecture as the oncogenic driver in this tumour type. To examine the epigenomic state of SyS we performed comprehensive multi-omics analysis on 52 primary pre-treatment human SyS tumours. Our analysis revealed a continuum of epigenomic states across the cohort at fusion target genes independent of rare somatic genetic lesions. We identify cell-of-origin signatures defined by enhancer states and reveal unexpected relationships between H2AK119Ub1 and active marks. The number of bivalent promoters, dually marked by the repressive H3K27me3 and activating H3K4me3 marks, has strong prognostic value and outperforms tumor grade in predicting patient outcome. Finally, we identify SyS defining epigenomic features including H3K4me3 expansion associated with striking promoter DNA hypomethylation in which SyS displays the lowest mean methylation level of any sarcoma subtype. We explore these distinctive features as potential vulnerabilities in SyS and identify H3K4me3 inhibition as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Jakob Hofvander
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Alvin Qiu
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, UBC, Vancouver, Canada
| | - Kiera Lee
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, UBC, Vancouver, Canada
| | - Misha Bilenky
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| | - Annaïck Carles
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Qi Cao
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Michelle Moksa
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Jonathan Steif
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Edmund Su
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
| | - Afroditi Sotiriou
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, Germany
- Soft-Tissue Sarcoma Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Germany
| | - Angela Goytain
- Department of Pathology and Laboratory Medicine, UBC, Vancouver, Canada
| | - Lesley A Hill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sam Singer
- Sarcoma Biology Laboratory, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene L Andrulis
- University Musculoskeletal Oncology Unit, Mount Sinai Hospital, Toronto, Canada
| | - Jay S Wunder
- Lunefeld-Tanenbaum Research Institute, Sinai Health System and Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Fredrik Mertens
- Division of Clinical Genetics, Lund University and Skåne University Hospital, Lund, Sweden
| | - Ana Banito
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, Germany
- Soft-Tissue Sarcoma Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kevin B Jones
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah, United States of America
- Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Torsten O Nielsen
- Department of Pathology and Laboratory Medicine, UBC, Vancouver, Canada
| | - Martin Hirst
- Department of Microbiology and Immunology, Michael Smith Laboratories, UBC, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| |
Collapse
|
12
|
Cheng B, Li L, Luo T, Wang Q, Luo Y, Bai S, Li K, Lai Y, Huang H. Single-cell deconvolution algorithms analysis unveils autocrine IL11-mediated resistance to docetaxel in prostate cancer via activation of the JAK1/STAT4 pathway. J Exp Clin Cancer Res 2024; 43:67. [PMID: 38429845 PMCID: PMC10905933 DOI: 10.1186/s13046-024-02962-8] [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: 11/20/2023] [Accepted: 01/19/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Docetaxel resistance represents a significant obstacle in the treatment of prostate cancer. The intricate interplay between cytokine signalling pathways and transcriptional control mechanisms in cancer cells contributes to chemotherapeutic resistance, yet the underlying molecular determinants remain only partially understood. This study elucidated a novel resistance mechanism mediated by the autocrine interaction of interleukin-11 (IL-11) and its receptor interleukin-11 receptor subunit alpha(IL-11RA), culminating in activation of the JAK1/STAT4 signalling axis and subsequent transcriptional upregulation of the oncogene c-MYC. METHODS Single-cell secretion profiling of prostate cancer organoid was analyzed to determine cytokine production profiles associated with docetaxel resistance.Analysis of the expression pattern of downstream receptor IL-11RA and enrichment of signal pathway to clarify the potential autocrine mechanism of IL-11.Next, chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) was performed to detect the nuclear localization and DNA-binding patterns of phosphorylated STAT4 (pSTAT4). Coimmunoprecipitation and reporter assays were utilized to assess interaction between pSTAT4 and the cotranscription factor CREB-binding protein (CBP) as well as their role in c-MYC transcriptional activity. RESULTS Autocrine secretion of IL-11 was markedly increased in docetaxel-resistant prostate cancer cells. IL-11 stimulation resulted in robust activation of JAK1/STAT4 signalling. Upon activation, pSTAT4 translocated to the nucleus and associated with CBP at the c-MYC promoter region, amplifying its transcriptional activity. Inhibition of the IL-11/IL-11RA interaction or disruption of the JAK1/STAT4 pathway significantly reduced pSTAT4 nuclear entry and its binding to CBP, leading to downregulation of c-MYC expression and restoration of docetaxel sensitivity. CONCLUSION Our findings identify an autocrine loop of IL-11/IL-11RA that confers docetaxel resistance through the JAK1/STAT4 pathway. The pSTAT4-CBP interaction serves as a critical enhancer of c-MYC transcriptional activity in prostate cancer cells. Targeting this signalling axis presents a potential therapeutic strategy to overcome docetaxel resistance in advanced prostate cancer.
Collapse
Affiliation(s)
- Bisheng Cheng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lingfeng Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Tianlong Luo
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 511430, China
| | - Yong Luo
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shoumin Bai
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Kaiwen Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yiming Lai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Hai Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|
13
|
Chang JY, Neugebauer C, Mues Genannt Koers A, 't Hart P. Small molecule WDR5 inhibitors down-regulate lncRNA expression. RSC Med Chem 2024; 15:636-640. [PMID: 38389889 PMCID: PMC10880924 DOI: 10.1039/d3md00605k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/10/2024] [Indexed: 02/24/2024] Open
Abstract
WD repeat domain 5 (WDR5) plays an important role as a scaffold protein in both protein-protein and RNA-protein complexes involved in epigenetic gene regulation. In particular, some of these lncRNAs were reported to regulate the expression of genes in cis as well as themselves through binding WDR5. In this report, we investigate the two known binding sites of WDR5 in relation to lncRNA binding and expression. The WBM binding site mediates both protein-protein and lncRNA-protein interactions while the WIN site, which is on the opposite side of the protein, is only known to mediate protein-protein interactions. To dissect the function of different binding sites on WDR5, we characterized them with selective peptide ligands using fluorescence polarization and used these to demonstrate the selectivity of small molecule inhibitors of these two major binding sites. RNA immunoprecipitation experiments were performed to show that lncRNA-WDR5 complex formation could be interrupted using a WBM site inhibitor. Finally, we demonstrated that WDR5 regulated lncRNAs are down regulated with different sensitivity toward the corresponding inhibitors, demonstrating the potential of targeting lncRNA-protein interactions to reduce oncogenic lncRNA expression.
Collapse
Affiliation(s)
- Jen-Yao Chang
- Chemical Genomics Centre of the Max Planck Society, Max Planck Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Cora Neugebauer
- Chemical Genomics Centre of the Max Planck Society, Max Planck Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Anne Mues Genannt Koers
- Chemical Genomics Centre of the Max Planck Society, Max Planck Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Peter 't Hart
- Chemical Genomics Centre of the Max Planck Society, Max Planck Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| |
Collapse
|
14
|
Chen Y, Huang M, Lu J, Zhang Q, Wu J, Peng S, Chen S, Zhang Y, Cheng L, Lin T, Chen X, Huang J. Establishment of a prognostic model to predict chemotherapy response and identification of RAC3 as a chemotherapeutic target in bladder cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:509-528. [PMID: 37310098 DOI: 10.1002/tox.23860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/17/2023] [Accepted: 05/28/2023] [Indexed: 06/14/2023]
Abstract
Cisplatin-based chemotherapy is considered the primary treatment option for patients with advanced bladder cancer (BCa). However, the objective response rate to chemotherapy is often unsatisfactory, leading to a poor 5-year survival rate. Furthermore, current strategies for evaluating chemotherapy response and prognosis are limited and inefficient. In this study, we aimed to address these challenges by establishing a chemotherapy response type gene (CRTG) signature consisting of 9 genes and verified the prognostic value of this signature using TCGA and GEO BCa cohorts. The risk scores based on the CRTG signature were found to be associated with advanced clinicopathological status and demonstrated favorable predictive power for chemotherapy response in the TCGA cohort. Meanwhile, tumors with high risk scores exhibited a tendency toward a "cold tumor" phenotype. These tumors showed a low abundance of T cells, CD8+ T cells and cytotoxic lymphocytes, along with a high abundance of cancer-associated fibroblasts. Moreover, they displayed higher mRNA levels of these immune checkpoints: CD200, CD276, CD44, NRP1, PDCD1LG2 (PD-L2), and TNFSF9. Furthermore, we developed a nomogram that integrated the CRTG signature with clinicopathologic risk factors. This nomogram proved to be a more effective tool for predicting the prognosis of BCa patients. Additionally, we identified Rac family small GTPase 3 (RAC3) as a biomarker in our model. RAC3 was found to be overexpressed in chemoresistant BCa tissues and enhance the chemotherapeutic resistance of BCa cells in vitro and in vivo by regulating the PAK1-ERK1/2 pathway. In conclusion, our study presents a novel CRTG model for predicting chemotherapy response and prognosis in BCa. We also highlight the potential of combining chemotherapy with immunotherapy as a promising strategy for chemoresistant BCa and that RAC3 might be a latent target for therapeutic intervention.
Collapse
Affiliation(s)
- Yuelong Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Ming Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Junlin Lu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Qiang Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Jilin Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Shengmeng Peng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Siting Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Yangjie Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Liang Cheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, PR China
| |
Collapse
|
15
|
Weissmiller AM, Fesik SW, Tansey WP. WD Repeat Domain 5 Inhibitors for Cancer Therapy: Not What You Think. J Clin Med 2024; 13:274. [PMID: 38202281 PMCID: PMC10779565 DOI: 10.3390/jcm13010274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
WDR5 is a conserved nuclear protein that scaffolds the assembly of epigenetic regulatory complexes and moonlights in functions ranging from recruiting MYC oncoproteins to chromatin to facilitating the integrity of mitosis. It is also a high-value target for anti-cancer therapies, with small molecule WDR5 inhibitors and degraders undergoing extensive preclinical assessment. WDR5 inhibitors were originally conceived as epigenetic modulators, proposed to inhibit cancer cells by reversing oncogenic patterns of histone H3 lysine 4 methylation-a notion that persists to this day. This premise, however, does not withstand contemporary inspection and establishes expectations for the mechanisms and utility of WDR5 inhibitors that can likely never be met. Here, we highlight salient misconceptions regarding WDR5 inhibitors as epigenetic modulators and provide a unified model for their action as a ribosome-directed anti-cancer therapy that helps focus understanding of when and how the tumor-inhibiting properties of these agents can best be understood and exploited.
Collapse
Affiliation(s)
- April M. Weissmiller
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 32132, USA;
| | - Stephen W. Fesik
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - William P. Tansey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA;
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
16
|
Tan Y, Qiao J, Yang S, Wang Q, Liu H, Liu Q, Feng W, Yang B, Li Z, Cui L. ARID5B-mediated LINC01128 epigenetically activated pyroptosis and apoptosis by promoting the formation of the BTF3/STAT3 complex in β2GPI/anti-β2GPI-treated monocytes. Clin Transl Med 2024; 14:e1539. [PMID: 38224186 PMCID: PMC10788880 DOI: 10.1002/ctm2.1539] [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: 09/17/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/16/2024] Open
Abstract
BACKGROUND Alterations of the trimethylation of histone 3 lysine 4 (H3K4me3) mark in monocytes are implicated in the development of autoimmune diseases. Therefore, the purpose of our study was to elucidate the role of H3K4me3-mediated epigenetics in the pathogenesis of antiphospholipid syndrome (APS). METHODS H3K4me3 Cleavage Under Targets and Tagmentation and Assay for Transposase-Accessible Chromatin were performed to determine the epigenetic profiles. Luciferase reporter assay, RNA immunoprecipitation, RNA pull-down, co-immunoprecipitation and chromatin immunoprecipitation were performed for mechanistic studies. Transmission electron microscopy and propidium iodide staining confirmed cell pyroptosis. Primary monocytes from patients with primary APS (PAPS) and healthy donors were utilised to test the levels of key molecules. A mouse model mimicked APS was constructed with beta2-glycoprotein I (β2GPI) injection. Blood velocity was detected using murine Doppler ultrasound. RESULTS H3K4me3 signal and open chromatin at the ARID5B promoter were increased in an in vitro model of APS. The epigenetic factor ARID5B directly activated LINC01128 transcription at its promoter. LINC01128 promoted the formation of the BTF3/STAT3 complex to enhance STAT3 phosphorylation. Activated STAT3 interacted with the NLRP3 promoter and subsequently stimulated pyroptosis and apoptosis. ARID5B or BTF3 depletion compensated for LINC01128-induced pyroptosis and apoptosis by inhibiting STAT3 phosphorylation. In mice with APS, β2GPI exposure elevated the levels of key proteins of pyroptosis and apoptosis pathways in bone marrow-derived monocytes, reduced the blood velocity of the ascending aorta, increased the thrombus size of the carotid artery, and promoted the release of interleukin (IL)-18, IL-1β and tissue factor. Patients with PAPS had the high-expressed ARID5B and LINC01128, especially those with triple positivity for antiphospholipid antibodies. Moreover, there was a positive correlation between ARID5B and LINC01128 expression. CONCLUSION This study indicated that ARID5B/LINC01128 was synergistically upregulated in APS, and they aggravated disease pathogenesis by enhancing the formation of the BTF3/STAT3 complex and boosting p-STAT3-mediated pyroptosis and apoptosis, thereby providing candidate therapeutic targets for APS. HIGHLIGHTS The H3K4me3 mark and chromatin accessibility at the ARID5B promoter are increased in vitro model mimicked APS. ARID5B-mediated LINC01128 induces pyroptosis and apoptosis via p-STAT3 by binding to BTF3. ARID5B is high- expressed in patients with primary APS and positively correlated with LINC01128 expression. OICR-9429 treatment mitigates pyroptosis and related inflammation in vivo and in vitro models mimicked APS.
Collapse
Affiliation(s)
- Yuan Tan
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Jiao Qiao
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Shuo Yang
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Qingchen Wang
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Hongchao Liu
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Qi Liu
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Weimin Feng
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Boxin Yang
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Zhongxin Li
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| | - Liyan Cui
- Institute of Medical TechnologyPeking University Health Science CenterBeijingChina
- Department of Laboratory MedicinePeking University Third HospitalBeijingChina
- Core Unit of National Clinical Research Center for Laboratory MedicinePeking University Third HospitalBeijingChina
| |
Collapse
|
17
|
Ogbuji V, Paster IC, Recio-Boiles A, Carew JS, Nawrocki ST, Chipollini J. Current Landscape of Immune Checkpoint Inhibitors for Metastatic Urothelial Carcinoma: Is There a Role for Additional T-Cell Blockade? Cancers (Basel) 2023; 16:131. [PMID: 38201559 PMCID: PMC10778285 DOI: 10.3390/cancers16010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/30/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Urothelial carcinoma (UC) is the most common form of bladder cancer (BC) and is the variant with the most immunogenic response. This makes urothelial carcinoma an ideal candidate for immunotherapy with immune checkpoint inhibitors. Key immune checkpoint proteins PD-1 and CTLA-4 are frequently expressed on T-cells in urothelial carcinoma. The blockade of this immune checkpoint can lead to the reactivation of lymphocytes and augment the anti-tumor immune response. The only immune checkpoint inhibitors that are FDA-approved for metastatic urothelial carcinoma target the programmed death-1 receptor and its ligand (PD-1/PD-L1) axis. However, the overall response rate and progression-free survival rates of these agents are limited in this patient population. Therefore, there is a need to find further immune-bolstering treatment combinations that may positively impact survival for patients with advanced UC. In this review, the current immune checkpoint inhibition treatment landscape is explored with an emphasis on combination therapy in the form of PD-1/PD-L1 with CTLA-4 blockade. The investigation of the current literature on immune checkpoint inhibition found that preclinical data show a decrease in tumor volumes and size when PD-1/PD-L1 is blocked, and similar results were observed with CTLA-4 blockade. However, there are limited investigations evaluating the combination of CTLA-4 and PD-1/PD-L1 blockade. We anticipate this review to provide a foundation for a deeper experimental investigation into combination immune checkpoint inhibition therapy in metastatic urothelial carcinoma.
Collapse
Affiliation(s)
- Vanessa Ogbuji
- College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (V.O.); (I.C.P.); (S.T.N.)
| | - Irasema C. Paster
- College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (V.O.); (I.C.P.); (S.T.N.)
| | - Alejandro Recio-Boiles
- Department of Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724, USA; (A.R.-B.); (J.S.C.)
| | - Jennifer S. Carew
- Department of Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724, USA; (A.R.-B.); (J.S.C.)
| | - Steffan T. Nawrocki
- College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (V.O.); (I.C.P.); (S.T.N.)
- Department of Medicine, The University of Arizona Cancer Center, Tucson, AZ 85724, USA; (A.R.-B.); (J.S.C.)
| | - Juan Chipollini
- College of Medicine, University of Arizona, Tucson, AZ 85724, USA; (V.O.); (I.C.P.); (S.T.N.)
| |
Collapse
|
18
|
Peng M, Chu X, Peng Y, Li D, Zhang Z, Wang W, Zhou X, Xiao D, Yang X. Targeted therapies in bladder cancer: signaling pathways, applications, and challenges. MedComm (Beijing) 2023; 4:e455. [PMID: 38107059 PMCID: PMC10724512 DOI: 10.1002/mco2.455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
Bladder cancer (BC) is one of the most prevalent malignancies in men. Understanding molecular characteristics via studying signaling pathways has made tremendous breakthroughs in BC therapies. Thus, targeted therapies including immune checkpoint inhibitors (ICIs), antibody-drug conjugates (ADCs), and tyrosine kinase inhibitor (TKI) have markedly improved advanced BC outcomes over the last few years. However, the considerable patients still progress after a period of treatment with current therapeutic regimens. Therefore, it is crucial to guide future drug development to improve BC survival, based on the molecular characteristics of BC and clinical outcomes of existing drugs. In this perspective, we summarize the applications and benefits of these targeted drugs and highlight our understanding of mechanisms of low response rates and immune escape of ICIs, ADCs toxicity, and TKI resistance. We also discuss potential solutions to these problems. In addition, we underscore the future drug development of targeting metabolic reprogramming and cancer stem cells (CSCs) with a deep understanding of their signaling pathways features. We expect that finding biomarkers, developing novo drugs and designing clinical trials with precisely selected patients and rationalized drugs will dramatically improve the quality of life and survival of patients with advanced BC.
Collapse
Affiliation(s)
- Mei Peng
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xuetong Chu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Yan Peng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Duo Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Zhirong Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Weifan Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xiaochen Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| |
Collapse
|
19
|
Hong X, Chen X, Wang H, Xu Q, Xiao K, Zhang Y, Chi Z, Liu Y, Liu G, Li H, Fang J, Lin T, Zhang Y. A HER2-targeted Antibody-Drug Conjugate, RC48-ADC, Exerted Promising Antitumor Efficacy and Safety with Intravesical Instillation in Preclinical Models of Bladder Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302377. [PMID: 37824205 PMCID: PMC10646285 DOI: 10.1002/advs.202302377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/30/2023] [Indexed: 10/14/2023]
Abstract
More than half of non-muscle-invasive bladder cancer (NMIBC) patients eventually relapse even if treated with surgery and BCG without optional bladder-preserving therapy. This study aims to investigate the antitumor activity and safety of a HER2-targeted antibody-drug conjugate, RC48-ADC, intravesical instillation for NMIBC treatment. In this preclinical study, it is revealed that human epidermal growth factor receptor 2 (HER2) expression scores of 1+, 2+, and 3+ are recorded for 16.7%, 56.2%, and 14.6% of NMIBC cases. The antitumor effect of RC48-ADC is positively correlated with HER2 expression in bladder cancer (BCa) cell lines and organoid models. Furthermore, RC48-ADC is revealed to exert its antitumor effect by inducing G2/M arrest and caspase-dependent apoptosis. In an orthotopic BCa model, tumor growth is significantly inhibited by intravesical instillation of RC48-ADC versus disitamab, monomethyl auristatin E, epirubicin, or phosphate-buffered saline control. The potential toxicity of intravesical RC48-ADC is also assessed by dose escalation in normal nude mice and revealed that administration of RC48-ADC by intravesical instillation is safe within the range of effective therapeutic doses. Taken together, RC48-ADC demonstrates promising antitumor effects and safety with intravesical administration in multiple preclinical models. These findings provide a rational for clinical trials of intravesical RC48-ADC in NMIBC patients.
Collapse
Affiliation(s)
- Xuwei Hong
- Department of UrologyShantou Central HospitalShantouGuangdong515031P. R. China
- Department of UrologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
| | - Xu Chen
- Department of UrologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
| | - Hongjin Wang
- Department of UrologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
| | - Qingchun Xu
- Department of UrologyShantou Central HospitalShantouGuangdong515031P. R. China
| | - Kanghua Xiao
- Department of UrologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
| | - Yuanfeng Zhang
- Department of UrologyShantou Central HospitalShantouGuangdong515031P. R. China
| | - Zepai Chi
- Department of UrologyShantou Central HospitalShantouGuangdong515031P. R. China
| | - Yeqing Liu
- Department of PathologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
| | - Guangyao Liu
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong510120P. R. China
| | - Hong Li
- BioMed LaboratoryGuangzhou Jingke Biotech GroupGuangzhouGuangdong510120P. R. China
| | - Jianmin Fang
- RemeGenLtd.YantaiShandong264006P. R. China
- School of Life Science and TechnologyTongji UniversityShanghai200092P. R. China
| | - Tianxin Lin
- Department of UrologySun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510120P. R. China
- Guangdong Provincial Clinical Research Centre for Urological DiseasesGuangzhouGuangdong510120P. R. China
| | - Yonghai Zhang
- Department of UrologyShantou Central HospitalShantouGuangdong515031P. R. China
| |
Collapse
|
20
|
Wang G, He X, Dai H, Lin L, Cao W, Fu Y, Diao W, Ding M, Zhang Q, Chen W, Guo H. WDR4 promotes the progression and lymphatic metastasis of bladder cancer via transcriptional down-regulation of ARRB2. Oncogenesis 2023; 12:47. [PMID: 37783676 PMCID: PMC10545698 DOI: 10.1038/s41389-023-00493-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/04/2023] Open
Abstract
Lymph node (LN) metastasis is one of the key prognostic factors in bladder cancer, but its underlying mechanisms remain unclear. Here, we found that elevated expression of WD repeat domain 4 (WDR4) in bladder cancer correlated with worse prognosis. WDR4 can promote the LN metastasis and proliferation of bladder cancer cells. Mechanistic studies showed that WDR4 can promote the nuclear localization of DEAD-box helicase 20 (DDX20) and act as an adaptor to bind DDX20 and Early growth response 1 (Egr1), thereby inhibiting Egr1-promoted transcriptional expression of arrestin beta 2 (ARRB2) and ultimately contributing to the progression of bladder cancer. Immunohistochemical analysis confirmed that WDR4 expression is also an independent predictor of LN metastasis in bladder cancer. Our results reveal a novel mechanism of LN metastasis and progression in bladder cancer and identify WDR4 as a potential therapeutic target for metastatic bladder cancer.
Collapse
Affiliation(s)
- Guoli Wang
- Department of Urology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Xin He
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Huiqi Dai
- Department of Urology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Lingyi Lin
- Department of Urology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Wenmin Cao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenli Diao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Meng Ding
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China
| | - Qing Zhang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China.
| | - Wei Chen
- Department of Urology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
21
|
Chen Y, Su H, Zhao J, Na Z, Jiang K, Bacchiocchi A, Loh KH, Halaban R, Wang Z, Cao X, Slavoff SA. Unannotated microprotein EMBOW regulates the interactome and chromatin and mitotic functions of WDR5. Cell Rep 2023; 42:113145. [PMID: 37725512 PMCID: PMC10629662 DOI: 10.1016/j.celrep.2023.113145] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
The conserved WD40-repeat protein WDR5 interacts with multiple proteins both inside and outside the nucleus. However, it is currently unclear whether and how the distribution of WDR5 between complexes is regulated. Here, we show that an unannotated microprotein EMBOW (endogenous microprotein binder of WDR5) dually encoded in the human SCRIB gene interacts with WDR5 and regulates its binding to multiple interaction partners, including KMT2A and KIF2A. EMBOW is cell cycle regulated, with two expression maxima at late G1 phase and G2/M phase. Loss of EMBOW decreases WDR5 interaction with KIF2A, aberrantly shortens mitotic spindle length, prolongs G2/M phase, and delays cell proliferation. In contrast, loss of EMBOW increases WDR5 interaction with KMT2A, leading to WDR5 binding to off-target genes, erroneously increasing H3K4me3 levels, and activating transcription of these genes. Together, these results implicate EMBOW as a regulator of WDR5 that regulates its interactions and prevents its off-target binding in multiple contexts.
Collapse
Affiliation(s)
- Yanran Chen
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Haomiao Su
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA
| | - Jianing Zhao
- Frontier Innovation Center, Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200433, China; Shanghai Fifth People's Hospital, Fudan University, Shanghai 200433, China
| | - Zhenkun Na
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA
| | - Kevin Jiang
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA
| | - Antonella Bacchiocchi
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ken H Loh
- Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ruth Halaban
- Department of Dermatology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Zhentian Wang
- Frontier Innovation Center, Department of Systems Biology for Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200433, China; Shanghai Fifth People's Hospital, Fudan University, Shanghai 200433, China
| | - Xiongwen Cao
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Sciences, East China Normal University, Shanghai 200062, China.
| | - Sarah A Slavoff
- Department of Chemistry, Yale University, New Haven, CT 06520, USA; Institute for Biomolecular Design and Discovery, Yale University, West Haven, CT 06516, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06529, USA.
| |
Collapse
|
22
|
Qiu L, Xu W, Lu X, Chen F, Chen Y, Tian Y, Zhu Q, Liu X, Wang Y, Pei XH, Xu X, Zhang J, Zhu WG. The HDAC6-RNF168 axis regulates H2A/H2A.X ubiquitination to enable double-strand break repair. Nucleic Acids Res 2023; 51:9166-9182. [PMID: 37503842 PMCID: PMC10516627 DOI: 10.1093/nar/gkad631] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/24/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
Histone deacetylase 6 (HDAC6) mediates DNA damage signaling by regulating the mismatch repair and nucleotide excision repair pathways. Whether HDAC6 also mediates DNA double-strand break (DSB) repair is unclear. Here, we report that HDAC6 negatively regulates DSB repair in an enzyme activity-independent manner. In unstressed cells, HDAC6 interacts with H2A/H2A.X to prevent its interaction with the E3 ligase RNF168. Upon sensing DSBs, RNF168 rapidly ubiquitinates HDAC6 at lysine 116, leading to HDAC6 proteasomal degradation and a restored interaction between RNF168 and H2A/H2A.X. H2A/H2A.X is ubiquitinated by RNF168, precipitating the recruitment of DSB repair factors (including 53BP1 and BRCA1) to chromatin and subsequent DNA repair. These findings reveal novel regulatory machinery based on an HDAC6-RNF168 axis that regulates the H2A/H2A.X ubiquitination status. Interfering with this axis might be leveraged to disrupt a key mechanism of cancer cell resistance to genotoxic damage and form a potential therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Lingyu Qiu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Wenchao Xu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xiaopeng Lu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Feng Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yongcan Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yuan Tian
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Qian Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xiangyu Liu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yongqing Wang
- Division of Rheumatology and Immunology, University of Toledo Medical Center, 3120 Glendale Avenue, Toledo 43614, OH, USA
| | - Xin-Hai Pei
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xingzhi Xu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Cell Biology and Medical Genetics, Shenzhen University Medical School, Shenzhen 518055, China
| | - Jun Zhang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
| | - Wei-Guo Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen 518055, China
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, China
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
23
|
Mouti MA, Deng S, Pook M, Malzahn J, Rendek A, Militi S, Nibhani R, Soonawalla Z, Oppermann U, Hwang CI, Pauklin S. KMT2A associates with PHF5A-PHF14-HMG20A-RAI1 subcomplex in pancreatic cancer stem cells and epigenetically regulates their characteristics. Nat Commun 2023; 14:5685. [PMID: 37709746 PMCID: PMC10502114 DOI: 10.1038/s41467-023-41297-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
Pancreatic cancer (PC), one of the most aggressive and life-threatening human malignancies, is known for its resistance to cytotoxic therapies. This is increasingly ascribed to the subpopulation of undifferentiated cells, known as pancreatic cancer stem cells (PCSCs), which display greater evolutionary fitness than other tumor cells to evade the cytotoxic effects of chemotherapy. PCSCs are crucial for tumor relapse as they possess 'stem cell-like' features that are characterized by self-renewal and differentiation. However, the molecular mechanisms that maintain the unique characteristics of PCSCs are poorly understood. Here, we identify the histone methyltransferase KMT2A as a physical binding partner of an RNA polymerase-associated PHF5A-PHF14-HMG20A-RAI1 protein subcomplex and an epigenetic regulator of PCSC properties and functions. Targeting the protein subcomplex in PCSCs with a KMT2A-WDR5 inhibitor attenuates their self-renewal capacity, cell viability, and in vivo tumorigenicity.
Collapse
Affiliation(s)
- Mai Abdel Mouti
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Siwei Deng
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Martin Pook
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Jessica Malzahn
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Aniko Rendek
- Department of Histopathology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Stefania Militi
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Reshma Nibhani
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Zahir Soonawalla
- Department of Hepatobiliary and Pancreatic Surgery, Oxford University Hospitals NHS, Oxford, UK
| | - Udo Oppermann
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Chang-Il Hwang
- Department of Microbiology and Molecular Genetics, University of California Davis, Davis, USA
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
24
|
Song Y, Peng Y, Qin C, Wang Y, Yang W, Du Y, Xu T. Fibroblast growth factor receptor 3 mutation attenuates response to immune checkpoint blockade in metastatic urothelial carcinoma by driving immunosuppressive microenvironment. J Immunother Cancer 2023; 11:e006643. [PMID: 37777251 PMCID: PMC10546120 DOI: 10.1136/jitc-2022-006643] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) therapy holds promise in metastatic urothelial carcinoma (UC). Fibroblast growth factor receptor 3 (FGFR3) mutation drives T-cell-depleted microenvironment in UC, which led to the hypothesis that FGFR3 mutation might attenuate response to ICB in patients with metastatic UC. The study aims to compare prognosis and response between patients with FGFR3-mutated and FGFR3-wildtype metastatic UC after ICB therapy, and decode the potential molecular mechanisms. METHODS Based on the single-arm, multicenter, phase 2 trial, IMvigor210, we conducted a propensity score matched (PSM) analysis. After a 1:1 ratio PSM method, 39 patients with FGFR3-mutated and 39 FGFR3-wildtype metastatic UC treated with atezolizumab were enrolled. A meta-analysis through systematical database retrieval was conducted for validation. In addition, we performed single-cell RNA sequencing on three FGFR3-mutated and three FGFR3-wildtype UC tumors and analyzed 58,069 single cells. RESULTS The PSM analysis indicated FGFR3-mutated patients had worse overall survival (OS) in comparison to FGFR3-wildtype patients (HR=2.11, 95% CI=(1.16 to 3.85), p=0.015) receiving atezolizumab. The median OS was 9.2 months (FGFR3-mutated) versus 21.0 months (FGFR3-wildtype). FGFR3-mutated patients had lower disease control rate than FGFR3-wildtype patients (41.0% vs 66.7%, p=0.023). The meta-analysis involving 938 patients with metastatic UC confirmed FGFR3 mutation was associated with worse OS after ICB (HR=1.28, 95% CI=(1.04 to 1.59), p=0.02). Single-cell RNA transcriptome analysis identified FGFR3-mutated UC carried a stronger immunosuppressive microenvironment compared with FGFR3-wildtype UC. FGFR3-mutated UC exhibited less immune infiltration, and lower T-cell cytotoxicity. Higher TREM2+ macrophage abundance in FGFR3-mutated UC can undermine and suppress the T cells, potentially contributing to the formation of an immunosuppressive microenvironment. Lower inflammatory-cancer-associated fibroblasts in FGFR3-mutated UC recruited less chemokines in antitumor immunity but expressed growth factors to promote FGFR3-mutated malignant cell development. FGFR3-mutated UC carried abundance of malignant cells characterized by high hypoxia/metabolism and low interferon response phenotype. CONCLUSIONS FGFR3 mutation can attenuate prognosis and response to ICB in patients with metastatic UC. FGFR3-mutated UC carries a stronger immunosuppressive microenvironment in comparison with FGFR3-wildtype UC. Inhibition of FGFR3 might activate the immune microenvironment, and the combination of FGFR inhibitor targeted therapy and ICB might be a promising therapeutic regimen in metastatic UC, providing important implications for UC clinical management.
Collapse
Affiliation(s)
- Yuxuan Song
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Yun Peng
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Caipeng Qin
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Yulong Wang
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Wenbo Yang
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Yiqing Du
- Department of Urology, Peking University People's Hospital, Beijing, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
25
|
Gurung R, Om D, Pun R, Hyun S, Shin D. Recent Progress in Modulation of WD40-Repeat Domain 5 Protein (WDR5): Inhibitors and Degraders. Cancers (Basel) 2023; 15:3910. [PMID: 37568727 PMCID: PMC10417795 DOI: 10.3390/cancers15153910] [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: 07/01/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
WD40-repeat (WDR) domain proteins play a crucial role in mediating protein-protein interactions that sustain oncogenesis in human cancers. One prominent example is the interaction between the transcription factor MYC and its chromatin co-factor, WD40-repeat domain protein 5 (WDR5), which is essential for oncogenic processes. The MYC family of proteins is frequently overexpressed in various cancers and has been validated as a promising target for anticancer therapies. The recruitment of MYC to chromatin is facilitated by WDR5, highlighting the significance of their interaction. Consequently, inhibiting the MYC-WDR5 interaction has been shown to induce the regression of malignant tumors, offering an alternative approach to targeting MYC in the development of anticancer drugs. WDR5 has two protein interaction sites, the "WDR5-binding motif" (WBM) site for MYC interaction and the histone methyltransferases SET1 recognition motif "WDR5-interacting" (WIN) site forming MLL complex. Significant efforts have been dedicated to the discovery of inhibitors that target the WDR5 protein. More recently, the successful application of targeted protein degradation technology has enabled the removal of WDR5. This breakthrough has opened up new avenues for inhibiting the interaction between WDR5 and the binding partners. In this review, we address the recent progress made in targeting WDR5 to inhibit MDR5-MYC and MDR5-MLL1 interactions, including its targeted protein degradation and their potential impact on anticancer drug discovery.
Collapse
Affiliation(s)
- Raju Gurung
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea; (R.G.); (D.O.); (R.P.)
| | - Darlami Om
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea; (R.G.); (D.O.); (R.P.)
| | - Rabin Pun
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea; (R.G.); (D.O.); (R.P.)
| | - Soonsil Hyun
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju-si 28160, Republic of Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea; (R.G.); (D.O.); (R.P.)
- Gachon Institute of Pharmaceutical Science, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| |
Collapse
|
26
|
Xiao K, Peng S, Lu J, Zhou T, Hong X, Chen S, Liu G, Li H, Huang J, Chen X, Lin T. UBE2S interacting with TRIM21 mediates the K11-linked ubiquitination of LPP to promote the lymphatic metastasis of bladder cancer. Cell Death Dis 2023; 14:408. [PMID: 37422473 PMCID: PMC10329682 DOI: 10.1038/s41419-023-05938-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/30/2023] [Indexed: 07/10/2023]
Abstract
Lymphatic metastasis is the most common pattern of bladder cancer (BCa) metastasis and has an extremely poor prognosis. Emerging evidence shows that ubiquitination plays crucial roles in various processes of tumors, including tumorigenesis and progression. However, the molecular mechanisms underlying the roles of ubiquitination in the lymphatic metastasis of BCa are largely unknown. In the present study, through bioinformatics analysis and validation in tissue samples, we found that the ubiquitin-conjugating E2 enzyme UBE2S was positively correlated with the lymphatic metastasis status, high tumor stage, histological grade, and poor prognosis of BCa patients. Functional assays showed that UBE2S promoted BCa cell migration and invasion in vitro, as well as lymphatic metastasis in vivo. Mechanistically, UBE2S interacted with tripartite motif containing 21 (TRIM21) and jointly induced the ubiquitination of lipoma preferred partner (LPP) via K11-linked polyubiquitination but not K48- or K63-linked polyubiquitination. Moreover, LPP silencing rescued the anti-metastatic phenotypes and inhibited the epithelial-mesenchymal transition of BCa cells after UBE2S knockdown. Finally, targeting UBE2S with cephalomannine distinctly inhibited the progression of BCa in cell lines and human BCa-derived organoids in vitro, as well as in a lymphatic metastasis model in vivo, without significant toxicity. In conclusion, our study reveals that UBE2S, by interacting with TRIM21, degrades LPP through K11-linked ubiquitination to promote the lymphatic metastasis of BCa, suggesting that UBE2S represents a potent and promising therapeutic target for metastatic BCa.
Collapse
Affiliation(s)
- Kanghua Xiao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China
| | - Shengmeng Peng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China
| | - Junlin Lu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China
| | - Ting Zhou
- Biobank of Sun Yat-sen University Cancer Center, Guangzhou, 510120, Guangdong, PR China
| | - Xuwei Hong
- Department of Urology, Shantou Central Hospital, Shantou, 515031, PR China
| | - Siting Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China
| | - Guangyao Liu
- School of Medicine, South China University of Technology, Guangzhou, 510120, Guangdong, PR China
| | - Hong Li
- BioMed Laboratory, Guangzhou Jingke Biotech Group, Guangzhou, 510120, Guangdong, PR China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510120, Guangdong, PR China.
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510120, Guangdong, PR China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, PR China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, Guangdong, PR China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510120, Guangdong, PR China.
| |
Collapse
|
27
|
Yao J, Liang Z, Duan L, G Y, Liu J, An G. Construction of a novel immune response prediction signature to predict the efficacy of immune checkpoint inhibitors in clear cell renal cell carcinoma patients. Heliyon 2023; 9:e15925. [PMID: 37484396 PMCID: PMC10360603 DOI: 10.1016/j.heliyon.2023.e15925] [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: 07/30/2022] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 07/25/2023] Open
Abstract
Background Immune checkpoint inhibitor (ICI) treatment has enhanced survival outcomes in clear cell renal cell carcinoma (ccRCC) patients. Nevertheless, the effectiveness of immunotherapy in ccRCC patients is restricted and we intended to develop and characterize an immune response prediction signature (IRPS) to forecast the efficacy of immunotherapy. Methods RNA-seq expression profile and clinicopathologic characteristics of 539 kidney cancer and 72 patients with normal specimens, were downloaded from the Cancer Genome Atlas (TCGA) database, while the Gene Expression Omnibus (GEO) database was used as the validation set, which included 24 ccRCC samples. Utilization of the TCGA data and immune genes databases (ImmPort and the InnateDB), we explored through Weighted Gene Co-expression Network Analysis (WGCNA), along with Least Absolute Shrinkage and Selection Operator method (LASSO), and constructed an IRPS for kidney cancer patients. GSEA and CIBERSORT were performed to declare the molecular and immunologic mechanism underlying the predictive value of IRPS. The Human Protein Atlas (HPA) was deployed to verify the protein expressions of IRPS genes. Tumor immune dysfunction and exclusion (TIDE) score and immunophenoscore (IPS) were computed to determine the risk of immune escape and value the discrimination of IRPS. A ccRCC cohort with anti-PD-1 therapy was obtained as an external validation data set to verify the predictive value of IRPS. Results We constructed a 10 gene signature related to the prognosis and immune response of ccRCC patients. Considering the IRPS risk score, patients were split into high and low risk groups. Patients with high risk in the TCGA cohort tended towards advanced tumor stage and grade with poor prognosis (p < 0.001), which was validated in GEO database (p = 0.004). High-risk group tumors were related with lower PD-L1 expression, higher TMB, higher MSIsensor score, lower IPS, higher TIDE score, and enriched Treg cells, which might be the potential mechanism of immune dysfunction and exclusion. Patients in the IRPS low risk group had better PFS (HR:0.73; 95% CI: 0.54-1.0; P = 0.047). Conclusion A novel biomarker of IRPS was constructed to predict the benefit of immunotherapy, which might lead to more individualized prognoses and tailored therapy for kidney cancer patients.
Collapse
Affiliation(s)
- Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ziwei Liang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ling Duan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yang G
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| |
Collapse
|
28
|
Ren D, Li L, Wang S, Zuo Y. The c-MYC transcription factor conduces to resistance to cisplatin by regulating MMS19 in bladder cancer cells. Tissue Cell 2023; 82:102096. [PMID: 37201439 DOI: 10.1016/j.tice.2023.102096] [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: 01/03/2023] [Revised: 04/09/2023] [Accepted: 04/15/2023] [Indexed: 05/20/2023]
Abstract
Chemoresistance is one of the dominant causes for tumor progression and recurrence of bladder cancer (BC). This paper investigated the effects of transcription factor c-MYC through promoting MMS19 expression on proliferation, metastasis and cisplatin (DDP) resistance in BC cells. The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database were applied to acquire the needed BC gene data. The mRNA and protein levels of c-MYC and MMS19 were verified with q-PCR or Western blot assay. MTT and Transwell assays were utilized to detect cell viability and metastasis. Chromatin Immunoprecipitation (ChIP) assay and Luciferase reporter assay were exerted to confirm the relationship between c-MYC and MMS19. TCGA and GEO BC datasets results implied MMS19 could be an independent indicator for BC patients' prognosis. MMS19 expression was dramatically augmented in BC cell lines. Overexpression of MMS19 conduced to accelerate BC cells proliferation, metastasis and increase DDP resistance. c-MYC was positively correlated with MMS19 and acted as a transcription activator for MMS19 in BC cell lines and activated MMS19 expression. Overexpression of c-MYC facilitated BC cells proliferation, metastasis and DDP resistance. In conclusions, c-MYC gene was a transcriptional regulator of MMS19. Up-regulation of c-MYC facilitated BC cells proliferation, metastasis and DDP resistance by motivating MMS19 expression. This molecular mechanism between c-MYC and MMS19 exerts a crucial mission in BC tumorigenesis and DDP resistance, and may contribute to the diagnosis and therapy of BC for the time to come.
Collapse
Affiliation(s)
- Da Ren
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Lei Li
- Department of Oncology, Binzhou Medical University Hospital, Binzhou 256603, Shandong Province, China
| | - Shuai Wang
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yali Zuo
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China.
| |
Collapse
|
29
|
Zhang Q, Liu S, Wang H, Xiao K, Lu J, Chen S, Huang M, Xie R, Lin T, Chen X. ETV4 Mediated Tumor-Associated Neutrophil Infiltration Facilitates Lymphangiogenesis and Lymphatic Metastasis of Bladder Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205613. [PMID: 36670069 PMCID: PMC10104629 DOI: 10.1002/advs.202205613] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/21/2022] [Indexed: 05/08/2023]
Abstract
As a key step of tumor lymphatic metastasis, lymphangiogenesis is regulated by VEGFC-VEGFR3 signaling pathway mediated by immune cells, mainly macrophages, in the tumor microenvironment. However, little is known whether tumor associated neutrophils are involved in lymphangiogenesis. Here, it is found that TANs infiltration is increased in LN-metastatic BCa and is associated with poor prognosis. Neutrophil depletion results in significant reduction in popliteal LN metastasis and lymphangiogenesis. Mechanistically, transcription factor ETV4 enhances BCa cells-derived CXCL1/8 to recruit TANs, leading to the increase of VEGFA and MMP9 from TANs, and then facilitating lymphangiogenesis and LN metastasis of BCa. Moreover, phosphorylation of ETV4 at tyrosine 392 by tyrosine kinase PTK6 increases nuclear translocation of ETV4 and is essential for its function in BCa. Overall, the findings reveal a novel mechanism of how tumor cells regulate TANs-induced lymphangiogenesis and LN metastasis and identify ETV4 as a therapeutic target of LN metastasis in BCa.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Sen Liu
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Hongjin Wang
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Kanghua Xiao
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Junlin Lu
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Siting Chen
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Ming Huang
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Ruihui Xie
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Tianxin Lin
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| | - Xu Chen
- Department of UrologySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouGuangdong510000P. R. China
- Guangdong Provincial Clinical Research Center for Urological DiseasesGuangzhouGuangdong510000P. R. China
| |
Collapse
|
30
|
Shah SD, Gillard BM, Wrobel MM, Karasik E, Moser MT, Mastri M, Long MD, Sule N, Brackett CM, Huss WJ, Foster BA. Syngeneic model of carcinogen-induced tumor mimics basal/squamous, stromal-rich, and neuroendocrine molecular and immunological features of muscle-invasive bladder cancer. Front Oncol 2023; 13:1120329. [PMID: 36816919 PMCID: PMC9936245 DOI: 10.3389/fonc.2023.1120329] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Bladder cancer is a heterogenous disease and the emerging knowledge on molecular classification of bladder tumors may impact treatment decisions based on molecular subtype. Pre-clinical models representing each subtype are needed to test novel therapies. Carcinogen-induced bladder cancer models represent heterogeneous, immune-competent, pre-clinical testing options with many features found in the human disease. Methods Invasive bladder tumors were induced in C57BL/6 mice when continuously exposed to N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) in the drinking water. Tumors were excised and serially passed by subcutaneous implantation into sex-matched syngeneic C57BL/6 hosts. Eight lines were named BBN-induced Urothelium Roswell Park (BURP) tumor lines. BURP lines were characterized by applying consensus molecular classification to RNA expression, histopathology, and immune profiles by CIBERSORT. Two lines were further characterized for cisplatin response. Results Eight BURP tumor lines were established with 3 male and 3 female BURP tumor lines, having the basal/squamous (BaSq) molecular phenotype and morphology. BURP-16SR was established from a male mouse and has a stromal-rich (SR) molecular phenotype and a sarcomatoid carcinoma morphology. BURP-19NE was established from a male mouse and has a neuroendocrine (NE)-like molecular phenotype and poorly differentiated morphology. The established BURP tumor lines have unique immune profiles with fewer immune infiltrates compared to their originating BBN-induced tumors. The immune profiles of the BURP tumor lines capture some of the features observed in the molecular classifications of human bladder cancer. BURP-16SR growth was inhibited by cisplatin treatment, while BURP-24BaSq did not respond to cisplatin. Discussion The BURP lines represent several molecular classifications, including basal/squamous, stroma-rich, and NE-like. The stroma-rich (BURP-16SR) and NE-like (BURP-19NE) represent unique immunocompetent models that can be used to test novel treatments in these less common bladder cancer subtypes. Six basal/squamous tumor lines were established from both male and female mice. Overall, the BURP tumor lines have less heterogeneity than the carcinogen-induced tumors and can be used to evaluate treatment response without the confounding mixed response often observed in heterogeneous tumors. Additionally, basal/squamous tumor lines were established and maintained in both male and female mice, thereby allowing these tumor lines to be used to compare differential treatment responses between sexes.
Collapse
Affiliation(s)
- Shruti D. Shah
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Bryan M. Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Michelle M. Wrobel
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Ellen Karasik
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Michael T. Moser
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Michalis Mastri
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Norbert Sule
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States,*Correspondence: Craig M. Brackett, ; Wendy J. Huss, ; Barbara A. Foster,
| | - Wendy J. Huss
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States,Department of Dermatology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States,*Correspondence: Craig M. Brackett, ; Wendy J. Huss, ; Barbara A. Foster,
| | - Barbara A. Foster
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States,*Correspondence: Craig M. Brackett, ; Wendy J. Huss, ; Barbara A. Foster,
| |
Collapse
|
31
|
Bianchi A, von Deimling M, Pallauf M, Yanagisawa T, Kawada T, Mostafaei H, Quhal F, Laukhtina E, Rajwa P, Majdoub M, Motlagh RS, Pradere B, Karakiewicz PI, Cerruto MA, Antonelli A, Shariat SF. Perspectives on the future of urothelial carcinoma therapy: chemotherapy and beyond. Expert Opin Pharmacother 2023; 24:177-195. [PMID: 36440477 DOI: 10.1080/14656566.2022.2150966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Despite recent developments in the landscape of urothelial carcinoma (UC) treatment, platinum combination chemotherapy still remains a milestone. Recently immunotherapeutic agents have gained ever-growing attractivity, particularly in the metastatic setting. Novel chemotherapeutic strategies and agents, such as antibody-drug conjugates (ADCs), and powerful combination regimens have been developed to overcome the resistance of most UC to current therapies. AREAS COVERED Herein, we review the current standard-of-care chemotherapy, the development of ADCs, the rationale for combining therapy regimens with chemotherapy in current trials, and future directions in UC management. EXPERT OPINION Immunotherapy has prompted a revolution in the treatment paradigm of UC. However, only a few patients experience a long-term response when treated with single-agent immunotherapies. Combination treatments are necessary to bypass resistance mechanisms and broaden the clinical utility of current options. Current evidence supports the intensification of standard-of-care chemotherapy with maintenance immunotherapy. However, the optimal sequence, combination, and duration must be determined to achieve individual longevity with acceptable health-related quality of life. In that regard, ADCs appear as a promising alternative for single and combination strategies in UC, as they specifically target the tumor cells, thereby, theoretically improving treatment efficacy and avoiding extensive off-target toxicities.
Collapse
Affiliation(s)
- Alberto Bianchi
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Markus von Deimling
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Pallauf
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, Paracelsus Medical University Salzburg, University Hospital Salzburg, Salzburg, Austria
| | - Takafumi Yanagisawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tatsushi Kawada
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hadi Mostafaei
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Research Center for Evidence Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fahad Quhal
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Pawel Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Muhammad Majdoub
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Reza Sari Motlagh
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Benjamin Pradere
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Department of Urology, La Croix Du Sud Hospital, Quint Fonsegrives, France
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, University of Montreal Health Centre, Montreal, Québec, Canada
| | - Maria Angela Cerruto
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Alessandro Antonelli
- Department of Urology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.,Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan.,Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Urology, Weill Cornell Medical College, New York, NY, USA.,Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria
| |
Collapse
|
32
|
Thomas P, Srivastava S, Udayashankara AH, Damodaran S, Yadav L, Mathew B, Suresh SB, Mandal AK, Srikantia N. RhoC in association with TET2/WDR5 regulates cancer stem cells by epigenetically modifying the expression of pluripotency genes. Cell Mol Life Sci 2022; 80:1. [PMID: 36469134 PMCID: PMC11073244 DOI: 10.1007/s00018-022-04645-z] [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: 08/25/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Emerging evidence illustrates that RhoC has divergent roles in cervical cancer progression where it controls epithelial to mesenchymal transition (EMT), migration, angiogenesis, invasion, tumor growth, and radiation response. Cancer stem cells (CSCs) are the primary cause of recurrence and metastasis and exhibit all of the above phenotypes. It, therefore, becomes imperative to understand if RhoC regulates CSCs in cervical cancer. In this study, cell lines and clinical specimen-based findings demonstrate that RhoC regulates tumor phenotypes such as clonogenicity and anoikis resistance. Accordingly, inhibition of RhoC abrogated these phenotypes. RNA-seq analysis revealed that RhoC over-expression resulted in up-regulation of 27% of the transcriptome. Further, the Infinium MethylationEPIC array showed that RhoC over-expressing cells had a demethylated genome. Studies divulged that RhoC via TET2 signaling regulated the demethylation of the genome. Further investigations comprising ChIP-seq, reporter assays, and mass spectrometry revealed that RhoC associates with WDR5 in the nucleus and regulates the expression of pluripotency genes such as Nanog. Interestingly, clinical specimen-based investigations revealed the existence of a subset of tumor cells marked by RhoC+/Nanog+ expression. Finally, combinatorial inhibition (in vitro) of RhoC and its partners (WDR5 and TET2) resulted in increased sensitization of clinical specimen-derived cells to radiation. These findings collectively reveal a novel role for nuclear RhoC in the epigenetic regulation of Nanog and identify RhoC as a regulator of CSCs. The study nominates RhoC and associated signaling pathways as therapeutic targets.
Collapse
Affiliation(s)
- Pavana Thomas
- Translational and Molecular Biology Laboratory (TMBL), Division of Molecular Biology and Genetics, St. John's Research Institute (SJRI), St. John's Medical College, Bangalore, 560034, India
- School of Integrative Health Sciences, The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - Sweta Srivastava
- Translational and Molecular Biology Laboratory (TMBL), Division of Molecular Biology and Genetics, St. John's Medical College Hospital, Bangalore, 560034, India.
| | - Avinash H Udayashankara
- Department of Radiation Oncology, St John's Medical College Hospital, Bangalore, 560034, India
| | - Samyuktha Damodaran
- Translational and Molecular Biology Laboratory (TMBL), Division of Molecular Biology and Genetics, St. John's Research Institute (SJRI), St. John's Medical College, Bangalore, 560034, India
| | - Lokendra Yadav
- Translational and Molecular Biology Laboratory (TMBL), Division of Molecular Biology and Genetics, St. John's Medical College Hospital, Bangalore, 560034, India
| | - Boby Mathew
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute (SJRI), St. John's Medical College, Bangalore, 560034, India
| | - Srinag Bangalore Suresh
- Translational and Molecular Biology Laboratory (TMBL), Division of Molecular Biology and Genetics, St. John's Research Institute (SJRI), St. John's Medical College, Bangalore, 560034, India
| | - Amit Kumar Mandal
- Clinical Proteomics Unit, Division of Molecular Medicine, St. John's Research Institute (SJRI), St. John's Medical College, Bangalore, 560034, India
| | - Nirmala Srikantia
- Department of Radiation Oncology, St John's Medical College Hospital, Bangalore, 560034, India
| |
Collapse
|
33
|
Wang J, Chen S, Wang H, Cao J, Fan X, Man J, Li Q, Yang L. Integrated molecular analyses of an interferon-γ based subtype with regard to outcome, immune characteristics, and immunotherapy in bladder cancer and experimental verification. Heliyon 2022; 8:e12102. [PMID: 36582677 PMCID: PMC9792807 DOI: 10.1016/j.heliyon.2022.e12102] [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: 08/31/2022] [Revised: 10/27/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
This study attempted to explore the role of interferon-γ related genes (IRGs) in the prognosis and immunotherapy of bladder cancer (BC). Based on data downloaded from public databases, molecular subtypes with different IRG expression patterns were determined via nonnegative matrix factorization clustering. On the basis of IRGs, interferon-γ related gene signature (IRGS) was developed through Cox regression analyses. We identified that two molecular subgroups with different outcome and immune profiles. It was proved that IRGS possessed prediction efficiency for BC prognosis. Compared with low IRGS group, high IRGS group was related to less anti-cancer immune cells infiltration, less tumor mutation burden score, more cancer stem cell index, and less benefit from immunotherapy. Differential expression of six model genes (IRF5, LATS2, MTHFD2, VAMP8, HLA-G and PTPN6) was validated between paired tissues by RT-qPCR. This study presents a prognostic model, which could serve as an indicator for the benefit of BC immunotherapy.
Collapse
Affiliation(s)
- Jirong Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Siyu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Huabin Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jinlong Cao
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Xinpeng Fan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Jiangwei Man
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Qingchao Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, Gansu, People's Republic of China
| |
Collapse
|
34
|
Li Z, Yang HY, Zhang XL, Zhang X, Huang YZ, Dai XY, Shi L, Zhou GR, Wei JF, Ding Q. Kinesin family member 23, regulated by FOXM1, promotes triple negative breast cancer progression via activating Wnt/β-catenin pathway. J Exp Clin Cancer Res 2022; 41:168. [PMID: 35524313 PMCID: PMC9077852 DOI: 10.1186/s13046-022-02373-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background Triple negative breast cancer (TNBC) is highly malignant and has a worse prognosis, compared with other subtypes of breast cancer due to the absence of therapeutic targets. KIF23 plays a crucial role in the tumorigenesis and cancer progression. However, the role of KIF23 in development of TNBC and the underlying mechanism remain unknown. The study aimed to elucidate the biological function and regulatory mechanism of KIF23 in TNBC. Methods Quantitative real-time PCR and Western blot were used to determine the KIF23 expression in breast cancer tissues and cell lines. Then, functional experiments in vitro and in vivo were performed to investigate the effects of KIF23 on tumor growth and metastasis in TNBC. Chromatin immunoprecipitation assay was conducted to illustrate the potential regulatory mechanisms of KIF23 in TNBC. Results We found that KIF23 was significantly up-regulated and associated with poor prognosis in TNBC. KIF23 could promote TNBC proliferation, migration and invasion in vitro and in vivo. KIF23 could activate Wnt/β-catenin pathway and promote EMT progression in TNBC. In addition, FOXM1, upregulated by WDR5 via H3K4me3 modification, directly bound to the promoter of KIF23 gene to promote its transcription and accelerated TNBC progression via Wnt/β-catenin pathway. Both of small inhibitor of FOXM1 and WDR5 could inhibit TNBC progression. Conclusions Our findings elucidate WDR5/FOXM1/KIF23/Wnt/β-catenin axis is associated with TNBC progression and may provide a novel and promising therapeutic target for TNBC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02373-7.
Collapse
|
35
|
Chen G, Chen Y, Xu R, Zhang G, Zou X, Wu G. Impact of SOX2 function and regulation on therapy resistance in bladder cancer. Front Oncol 2022; 12:1020675. [PMID: 36465380 PMCID: PMC9709205 DOI: 10.3389/fonc.2022.1020675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2024] Open
Abstract
Bladder cancer (BC) is a malignant disease with high rates of recurrence and mortality. It is mainly classified as non-muscle-invasive BC and muscle-invasive BC (MIBC). Often, MIBC is chemoresistant, which, according to cancer stem cells (CSCs) theory, is linked to the presence of bladder cancer stem cells (BCSCs). Sex-determining region Y- (SRY) Box transcription factor 2 (SOX2), which is a molecular marker of BCSCs, is aberrantly over-expressed in chemoresistant BC cell lines. It is one of the standalone prognostic factors for BC, and it has an inherently significant function in the emergence and progression of the disease. This review first summarizes the role of SRY-related high-mobility group protein Box (SOX) family genes in BC, focusing on the SOX2 and its significance in BC. Second, it discusses the mechanisms relevant to the regulation of SOX2. Finally, it summarizes the signaling pathways related to SOX2 in BC, suggests current issues to be addressed, and proposes potential directions for future research to provide new insights for the treatment of BC.
Collapse
Affiliation(s)
- Guodong Chen
- The First Clinical College, Gannan Medical University, Ganzhou, China
- Department of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yan Chen
- Department of Gastroenterology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruiquan Xu
- Department of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Guoxi Zhang
- Department of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Gengqing Wu
- Department of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
36
|
Tang L, Peng L, Tan C, Liu H, Chen P, Wang H. Role of HOXA9 in solid tumors: mechanistic insights and therapeutic potential. Cancer Cell Int 2022; 22:349. [PMID: 36376832 PMCID: PMC9664671 DOI: 10.1186/s12935-022-02767-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
HOXA9 functioning as a transcription factor is one of the members of HOX gene family, which governs multiple cellular activities by facilitating cellular signal transduction. In addition to be a driver in AML which has been widely studied, the role of HOXA9 in solid tumor progression has also received increasing attention in recent years, where the aberrant expression of HOXA9 is closely associated with the prognosis of patient. This review details the signaling pathways, binding partners, post-transcriptional regulation of HOXA9, and possible inhibitors of HOXA9 in solid tumors, which provides a reference basis for further study on the role of HOXA9 in solid tumors.
Collapse
|
37
|
Zou Y, Yuan G, Tan X, Luo S, Yang C, Tang Y, Wang Y, Yao K. Immune-related gene risk score predicting the effect of immunotherapy and prognosis in bladder cancer patients. Front Genet 2022; 13:1011390. [PMID: 36267410 PMCID: PMC9577248 DOI: 10.3389/fgene.2022.1011390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Immune checkpoint inhibitor therapy has changed the treatment model of metastatic bladder cancer. However, only approximately 20% of patients benefit from this therapy, and robust biomarkers to predict the effect of immunotherapy are still lacking. In this study, we aimed to investigate whether immune-related genes could be indicators for the prognosis of bladder cancer patients and the effect of immunotherapy. Methods: Based on bladder cancer dataset from the Cancer Genome Atlas (TCGA) and GSE48075, 22 immune microenvironment-related cells were identified by CIBERSORT. After performing a series of bioinformatic and machine learning approaches, we identified distinct tumor microenvironment clusters and three bladder cancer specific immune-related genes (EGFR, OAS1 and MST1R). Then, we constructed immune-related gene risk score (IRGRS) by using the Cox regression method and validated it with the IMvigor210 dataset. Results: IRGRS-high patients had a worse overall survival than IRGRS-low patients, which was consistent with the result in the IMvigor210 dataset. Comprehensive analysis shows that patients with high IRGRS scores are mainly enriched in basal/squamous type (Ba/Sq), and tumor metabolism-related pathways are more Active, with higher TP53 and RB1 gene mutation rates, lower CD4+/CD8+ T cell infiltration, higher M0 macrophage infiltration, and lower immunotherapy efficacy. In contrast, Patients with low IRGRS scores are mainly enriched in the luminal papillary type (LumP), which is associated with the activation of IL-17 and TNF signaling pathways, higher mutation rates of FGFR3 and CDKN1A genes, higher CD4+/CD8+ T cell infiltration content, and The level of M0 macrophage infiltration was relatively low, and the immunotherapy was more probably effective. Conclusion: Our study constructed an IRGRS for bladder cancer and clarified the immune and molecular characteristics of IRGRS-defined subgroups of bladder cancer to investigate the association between IRGRS and its potential implications for prognosis and immunotherapy.
Collapse
Affiliation(s)
- Yuantao Zou
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Gangjun Yuan
- Department of Urology Oncological Surgery, Chongqing University Cancer Hospital, Chongqing, China
| | - Xingliang Tan
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Sihao Luo
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Cong Yang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yi Tang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yanjun Wang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Kai Yao
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in Southern China, Guangzhou, China
- Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
- *Correspondence: Kai Yao,
| |
Collapse
|
38
|
Song Z, Li H. Circ_0072995 drives cervical cancer development by regulating the miR-29a/WDR5 axis. J OBSTET GYNAECOL 2022; 42:3342-3348. [PMID: 36168936 DOI: 10.1080/01443615.2022.2125294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Circular RNA_0072995 (Circ_0072995) is involved in the pathogenesis of cancers; however, no study has investigated its role in cervical cancer. Therefore, we aimed to investigate the function of circ_0072995 in cervical cancer. Normal human cervical epithelial cells (hCECs), HeLa cells, and forty female nude BALB/c mice were used. Immunohistochemistry, invasion assays, flow cytometric analysis, luciferase assays, and tumour volume measurements were performed to explore the potential mechanism. Circ_0072995 was significantly up-regulated in cancer tissues, and its level was markedly correlated with the International Federation of Gynecology and Obstetrics system (FIGO) staging. In vitro studies revealed that circ_0072995 interacts with miR-29a to induce WD repeat domain 5 (WDR5) expression and promotes the proliferation and invasion of cells, but inhibits apoptosis of cells. Knockdown of circ_0072995 or WDR5, or overexpression of miR-29a significantly inhibited tumour growth in vivo. In conclusion, circ_0072995 promoted cervical cancer development by inducing miR-29a-mediated WDR5 expression.Impact statementWhat is already known on this subject? Global Cancer Statistics 2020 estimated that there were 1 021,494 new cases of cervical cancer and 439 201 deaths from cervical cancer. Circ_0072995 was first shown to promote breast cancer development in 2018. Subsequent studies have revealed that circ_0072995 is also involved in the development of other cancers, including epithelial ovarian cancer and hepatocellular carcinoma. However, no studies have explored the association between circ_0072995 and cervical cancer.What do the results of this study add? We hypothesized that circ_0072995 drives cervical cancer development by sponging miRNAs and inducing the expression of key factors involved in tumorigenesis. Based on this hypothesis, we investigated the role of circ_0072995 in cervical cancer and paracancerous tissues and explored the underlying mechanism in both in vitro and in vivo studies.What are the implications of these findings for clinical practice and/or further research? For the first time, our study revealed the key role of WDR5 in cervical cancer progression regulated by circ_0072995. We first reported the promoting effects of circ_0072995 in cervical cancer development by inducing miR-29a mediated WDR5 expression and also revealed the therapeutic potential of circ_0072995, miR-29a, and WDR5 in cervical cancer.
Collapse
Affiliation(s)
- Zhenzhen Song
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Wang K, Dai X, Yu A, Feng C, Liu K, Huang L. Peptide-based PROTAC degrader of FOXM1 suppresses cancer and decreases GLUT1 and PD-L1 expression. J Exp Clin Cancer Res 2022; 41:289. [PMID: 36171633 PMCID: PMC9520815 DOI: 10.1186/s13046-022-02483-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background Peptide proteolysis-targeting chimeras (p-PROTACs) with advantages of high specificity and low toxicity have emerged as a powerful technology of targeted protein degradation for biomedical applications. FOXM1, a proliferation-associated transcription factor, is overexpressed in a variety of human tumors as a key driver of tumorigenesis and cancer progression, and is a potential anticancer therapeutic target. However, FOXM1-targeting p-PROTACs has not been researched. Methods Here, we first analyzed the expression of FOXM1, GLUT1 and PD-L1 in liver cancer through database and clinical samples of patients. FOXM1-targeting peptides, selected by screening phage display library, are verified its targeting effect by immunofluorescence and CCK-8 test. The novel p-PROTAC degrader of FOXM1 is chemically synthesis, named FOXM1-PROTAC, by linking a FOXM1-binding antagonistic peptide, with the E3 ubiquitin ligase recruitment ligand Pomalidomide and with the cell membrane penetrating peptide TAT. Its degradation effect on FOXM1 was detected by Western blotting, qPCR, and we verified its effect on the behavior of cancer cells by flow cytometry, scratch assay, and Transwell in vitro. The tumor xenografted mice model was used for evaluating FOXM1-PROTAC therapeutic response in vivo. Finally, we detected the expression of GLUT1 and PD-L1 after FOXM1-PROTAC degraded FOXM1 by using Western Blotting and hippocampal detectors and dual immunofluorescence. Results We found that the novel FOXM1-PROTAC efficiently entered cells and induced degradation of FOXM1 protein, which strongly inhibits viability as well as migration and invasion in various cancer cell lines, and suppressed tumor growth in HepG2 and MDA-MB-231 cells xenograft mouse models, without detected toxicity in normal tissues. Meanwhile, FOXM1-PROTAC decreased the cancer cells glucose metabolism via downregulating the protein expression levels of glucose transporter GLUT1 and the immune checkpoint PD-L1, which suggests involvement of FOXM1 in cancer cell metabolism and immune regulation. Conclusions Our results indicate that biologically targeted degradation of FOXM1 is an attractive therapeutic strategy, and antagonist peptide-containing FOXM1-PROTACs as both degrader and inhibitor of FOXM1 could be developed as a safe and promising drug for FOXM1-overexpressed cancer therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02483-2.
Collapse
|
40
|
He Z, Gu J, Luan T, Li H, Li C, Chen Z, Luo E, Wang J, Huang Y, Ding M. Comprehensive analyses of a tumor-infiltrating lymphocytes-related gene signature regarding the prognosis and immunologic features for immunotherapy in bladder cancer on the basis of WGCNA. Front Immunol 2022; 13:973974. [PMID: 36211333 PMCID: PMC9540212 DOI: 10.3389/fimmu.2022.973974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor-infiltrating lymphocyte (TIL) is a class of cells with important immune functions and plays a crucial role in bladder cancer (BCa). Several studies have shown the clinical significance of TIL in predicting the prognosis and immunotherapy efficacy. TIL-related gene module was screened utilizing weighted gene coexpression network analysis. We screened eight TIL-related genes utilizing univariate Cox regression analysis, least absolute shrinkage and selection operator (LASSO) Cox regression analysis, and multivariate Cox regression analysis. Then, we established a TIL-related signature model containing the eight selected genes and subsequently classified all patients into two groups, that is, the high-risk as well as low-risk groups. Gene mutation status, prognosis, immune cell infiltration, immune subtypes, TME, clinical features, and immunotherapy response were assessed among different risk subgroups. The results affirmed that the TIL-related signature model was a reliable predictor of overall survival (OS) for BCa and was determined as an independent risk factor for BCa patients in two cohorts. Moreover, the risk score was substantially linked to age, tumor staging, TNM stage, and pathological grade. And there were different mutational profiles, biological pathways, immune scores, stromal scores, and immune cell infiltration in the tumor microenvironment (TME) between the two risk groups. In particular, immune checkpoint genes’ expression was remarkably different between the two risk groups, with patients belonging to the low-risk group responding better to immune checkpoint inhibition (ICI) therapy. In conclusion, our study demonstrates that the TIL-related model was a reliable signature in anticipating prognosis, immune status, and immunotherapy response, which can help in screening patients who respond to immunotherapy.
Collapse
Affiliation(s)
- Zexi He
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Gu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ting Luan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haihao Li
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Charles Li
- Zhongke Jianlan Medical Research Institute, Beijing, China
| | - Zhenjie Chen
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Enxiu Luo
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yinglong Huang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Mingxia Ding, ; Yinglong Huang,
| | - Mingxia Ding
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Urological Disease Clinical Medical Center of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Mingxia Ding, ; Yinglong Huang,
| |
Collapse
|
41
|
Rad54L promotes bladder cancer progression by regulating cell cycle and cell senescence. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:185. [PMID: 36071250 DOI: 10.1007/s12032-022-01751-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/13/2022] [Indexed: 10/14/2022]
Abstract
Bladder cancer (BCa) is the most prevalent cancer of the urinary system, but its pathogenesis is still poorly understood. Several reports have suggested that gene damage repair is highly correlated with tumor development and drug resistance, in which homologous recombination repair gene Rad54L seems to play an important role, through yet unclear mechanisms. Therefore, this study stratified cancer patients by Rad54L expression in BCa tissue, and high Rad54L expression was associated with a poor prognosis. Mechanistically, we demonstrate that high Rad54L expression promotes abnormal bladder tumor cell proliferation by changing the cell cycle and cell senescence. In addition, this study also suggests that Rad54L may be associated with p53, p21, and pRB in BCa tissue. In summary, this study exposes Rad54L as potential a prognostic biomarker and precision treatment target in BCa.
Collapse
|
42
|
Ma T, Gu J, Wen H, Xu F, Ge D. BIRC5 Modulates PD-L1 Expression and Immune Infiltration in Lung Adenocarcinoma. J Cancer 2022; 13:3140-3150. [PMID: 36046648 PMCID: PMC9414029 DOI: 10.7150/jca.69236] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
Background: Lung adenocarcinoma (LUAD) is the most prevalent thoracic cancer with the highest incidence and mortality worldwide. Baculoviral IAP Repeat Containing 5 (BIRC5) is well studied in many malignancies, its prognosis value and correlation with the tumor microenvironment (TME) in LAUD remains largely elusive. Methods: The Wilcoxon signed-rank test and logistic regression were used to evaluate the relationship between clinical features and BIRC5 expression in LUAD. To assess the impact of BIRC5 on prognosis, the Kaplan-Meier plotter analysis and Cox regression were used, as well as a receiver operating characteristic (ROC) curve and nomogram. Gene set enrichment analysis (GSEA) and single-sample gene set enrichment analysis (ssGSEA) were recruited to predict the association between BIRC5 and immune cell infiltrations. Furthermore, qRT-PCR and western bolt were utilized to confirm gene expression on mRNA and protein levels. The proliferation of A549 and H1299 cells was evaluated using CCK8 and EdU assay. Cell mobility was tested by transwell assay and wound healing assay. Detection of PD-L1 and infiltrated CD8 T cells in xenograft tumors was done by flow cytometry. Results: BIRC5 expression was found to be substantially greater in LUAD patients. According to KM-plotter analysis, patients with high levels of BIRC5 had shorter survival rates. Multivariate Cox analysis revealed that elevated BIRC5 expression was an independent risk factor for OS and PFS in LUAD patients. High BIRC5 expression was predicted to be associated with chemokine activity and immune cell chemotaxis, whereas ssGSEA suggested that BIRC5 is highly associated with CD8 T cell infiltration and PD-L1 levels. In vitro experiments suggested overexpression of BIRC5 promoted the proliferation, mobility, and PD-L1 level of A549 cells, and vice versa in H1299 cells. Furthermore, in vivo study suggested elevated tumor weight and PD-L1 levels in xenograft tumors generated from LLC cells with overexpressed BIRC5. Conclusion: BIRC5 promotes lung adenocarcinoma progression by modulating PD-L1 expression and inducing tumor immune evasion.
Collapse
Affiliation(s)
- Teng Ma
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Gu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haoyu Wen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fengkai Xu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
43
|
Zhang Z, Han Y, Sun Q, Wang Y, Sun L. The DPY30-H3K4me3 Axis-Mediated PD-L1 Expression in Melanoma. J Inflamm Res 2022; 15:5595-5609. [PMID: 36185638 PMCID: PMC9525212 DOI: 10.2147/jir.s377678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background DPY30 is a common subunit of the human SET1/MLL complex and is an essential protein required for the activity of SET1/MLL methyltransferase. DPY30 regulates the histone H3K4 modification, and dysfunction of DPY30 might contribute to the regulation of cancer immune evasion. However, the functions and regulation of DPY30 in the expression of programmed cell death ligand 1 (PD-L1) is still not completely explored. Methods Various online databases were used for data processing and visualization, including UALCAN, Oncomine, cBioPortal, SangerBox, TISIDB, TIMER, and GEPIA databases. The expression of DPY30 and PD-L1 in melanoma tissues were evaluated by IHC. Chromatin Immunoprecipitation (ChIP), RT-PCR and flow cytometry were used to elucidate the underlying molecular mechanism of PD-L1 expression regulation and its function. Results The mRNA level of DPY30 in melanoma was higher than in normal tissues. The expression of DPY30 was positively associated with TMB, neoantigens and PD-L1 expression. Furthermore, DPY30 expression showed significant positive correlations with immune suppressor cells and ICP genes involved in T-cell exhaustion. IHC showed that the positive rates of DPY30 and PD-L1 in melanoma tissues were 62% and 58%, respectively. Correlation analysis revealed that DPY30 over-expression was positively associated with PD-L1 expression. Silencing of DPY30 by specific siRNA significantly inhibited PD-L1 expression. ChIP analysis revealed that H3K4me3 levels were enriched in the proximal PD-L1 promoter region in tumor cells. Inhibition of DPY30 still suppressed the PD-L1 level in IFN-γ treated MMAC-SF cells. Furthermore, the apoptosis of PD1+ T-cells in co-culture with MMAC-SF cells by knockdown of DPY30 were markedly reduced. Conclusion This study shows the roles of DPY30 in regulating the cancer immune evasion in melanoma. Targeting the DPY30-H3K4me3 axis might be an alternative approach to enhance the efficacy of checkpoint immunotherapy.
Collapse
Affiliation(s)
- Zhichun Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yixuan Han
- Department of Rheumatology and Immunology, Affiliated Kailuan General Hospital of North China University of Science and Technology, Tangshan, People’s Republic of China
| | - Qiuyue Sun
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Yipeng Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Lichao Sun
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People’s Republic of China
- Correspondence: Lichao Sun; Yipeng Wang, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China, Tel/Fax +86 10-67781331, Email ;
| |
Collapse
|
44
|
A novel molecular subtypes and risk model based on inflammatory response-related lncrnas for bladder cancer. Hereditas 2022; 159:32. [PMID: 35964079 PMCID: PMC9375404 DOI: 10.1186/s41065-022-00245-w] [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: 04/22/2022] [Accepted: 07/28/2022] [Indexed: 12/24/2022] Open
Abstract
Background Inflammation and long noncoding RNAs (lncRNAs) are gradually becoming important in the development of bladder cancer (BC). Nevertheless, the potential of inflammatory response-related lncRNAs (IRRlncRNAs) as a prognostic signature remains unexplored in BC. Methods The Cancer Genome Atlas (TCGA) provided RNA expression profiles and clinical information of BC samples, and GSEA Molecular Signatures database provided 1171 inflammation-related genes. IRRlncRNAs were identified using Pearson correlation analysis. After that, consensus clustering was performed to form molecular subtypes. After performing least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analyses, a risk model constructed based on the prognostic IRRlncRNAs was validated in an independent cohort. Kaplan–Meier (KM) analysis, univariate and multivariate Cox regression, clinical stratification analysis, and time-dependent receiver operating characteristic (ROC) curves were utilized to assess clinical effectiveness and accuracy of the risk model. In clusters and risk model, functional enrichment was investigated using GSEA and GSVA, and immune cell infiltration analysis was demonstrated by ESTIMATE and CIBERSORT analysis. Results A total of 174 prognostic IRRlncRNAs were confirmed, and 406 samples were divided into 2 clusters, with cluster 2 having a significantly inferior prognosis. Moreover, cluster 2 exhibited a higher ESTIMATE score, immune infiltration, and PD-L1 expression, with close relationships with the inflammatory response. Further, 12 IRRlncRNAs were identified and applied to construct the risk model and divide BC samples into low-risk and high-risk groups successfully. KM, ROC, and clinical stratification analysis demonstrated that the risk model performed well in predicting prognosis. The risk score was identified as an independently significant indicator, enriched in immune, cell cycle, and apoptosis-related pathways, and correlated with 9 immune cells. Conclusion We developed an inflammatory response-related subtypes and steady prognostic risk model based on 12 IRRlncRNAs, which was valuable for individual prognostic prediction and stratification and outfitted new insight into inflammatory response in BC. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-022-00245-w.
Collapse
|
45
|
Sun JX, Liu CQ, Xu JZ, An Y, Xu MY, Zhong XY, Zeng N, Ma SY, He HD, Zhang ZB, Wang SG, Xia QD. A Four-Cell-Senescence-Regulator-Gene Prognostic Index Verified by Genome-Wide CRISPR Can Depict the Tumor Microenvironment and Guide Clinical Treatment of Bladder Cancer. Front Immunol 2022; 13:908068. [PMID: 35898492 PMCID: PMC9312376 DOI: 10.3389/fimmu.2022.908068] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/08/2022] [Indexed: 01/10/2023] Open
Abstract
Bladder cancer (BCa) is the 10th most commonly diagnosed cancer worldwide, and cellular senescence is defined as a state of permanent cell cycle arrest and considered to play important roles in the development and progression of tumor. However, the comprehensive effect of senescence in BCa has not ever been systematically evaluated. Using the genome-wide CRISPR screening data acquired from DepMap (Cancer Dependency Map), senescence genes from the CellAge database, and gene expression data from The Cancer Genome Atlas (TCGA), we screened out 12 senescence genes which might play critical roles in BCa. A four-cell-senescence-regulator-gene prognostic index was constructed using the least absolute shrinkage and selection operator (LASSO) and multivariate COX regression model. The transcriptomic data and clinical information of BCa patients were downloaded from TCGA and Gene Expression Omnibus (GEO). We randomly divided the patients in TCGA cohort into training and testing cohorts and calculated the risk score according to the expression of the four senescence genes. The validity of this risk score was validated in the testing cohort (TCGA) and validation cohort (GSE13507). The Kaplan–Meier curves revealed a significant difference in the survival outcome between the high- and low-risk score groups. A nomogram including the risk score and other clinical factors (age, gender, stage, and grade) was established with better predictive capacity of OS in 1, 3, and 5 years. Besides, we found that patients in the high-risk group had higher tumor mutation burden (TMB); lower immune, stroma, and ESTIMATE scores; higher tumor purity; aberrant immune functions; and lower expression of immune checkpoints. We also performed gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA) to investigate the interaction between risk score and hallmark pathways and found that a high risk score was connected with activation of senescence-related pathways. Furthermore, we found that a high risk score was related to better response to immunotherapy and chemotherapy. In conclusion, we identified a four-cell-senescence-regulator-gene prognostic index in BCa and investigated its relationship with TMB, the immune landscape of tumor microenvironment (TME), and response to immunotherapy and chemotherapy, and we also established a nomogram to predict the prognosis of patients with BCa.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zong-Biao Zhang
- *Correspondence: Zong-Biao Zhang, ; Shao-Gang Wang, ; Qi-Dong Xia,
| | - Shao-Gang Wang
- *Correspondence: Zong-Biao Zhang, ; Shao-Gang Wang, ; Qi-Dong Xia,
| | - Qi-Dong Xia
- *Correspondence: Zong-Biao Zhang, ; Shao-Gang Wang, ; Qi-Dong Xia,
| |
Collapse
|
46
|
Zhang X, Tian C, Tian C, Cheng J, Mao W, Li M, Chen M. LTBP2 inhibits prostate cancer progression and metastasis via the PI3K/AKT signaling pathway. Exp Ther Med 2022; 24:563. [PMID: 36034756 PMCID: PMC9400130 DOI: 10.3892/etm.2022.11500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/11/2022] [Indexed: 12/02/2022] Open
Abstract
Biochemical recurrence (BCR) is a cause of concern in advanced prostate cancer (PCa). Thus, novel diagnostic biomarkers are required to improve clinical care. However, research on PCa immunotherapy is also scarce. Hence, the present study aimed to explore promising BCR-related diagnostic biomarkers, and their expression pattern, prognostic value, immune response effects, biological functions, and possible molecular mechanisms were evaluated. GEO datasets (GSE46602, GSE70768, and GSE116918) were downloaded and merged as the training cohort, and differential expression analysis was performed. Lasso regression and SVM-RFE algorithm, as well as PPI analysis and MCODE algorithm, were then applied to filter BCR-related biomarker genes. The CIBERSORT and estimation of stromal and immune cells in malignant tumor tissues using expression data (ESTIMATE) methods were used to calculate the fractions of tumor-infiltrating immune cells. GO/DO enrichment analyses were used to identify the biological functions. The expression of latent transforming growth factor β-binding protein 2 (LTBP2) was determined by RT-qPCR and western blotting. The role of LTBP2 in PCa was determined by CCK-8, Transwell, and the potential mechanism was investigated by KEGG and GSEA and confirmed by western blotting. In total, 44 BCR-related differentially expressed genes (DEGs) in the training cohort were screened. LTBP2 was found to be a diagnostic biomarker of BCR in PCa and was associated with CD4+ T-cell infiltration and response to anti-PD-1/PD-L1 immunotherapy. Subsequently, using the ESTIMATE algorithm, it was identified that LTBP2 was associated with the tumor microenvironment and could be a predictor of the clinical benefit of immune checkpoint blockade. Finally, the expression and biological function of LTBP2 were evaluated via cellular experiments. The results showed that LTBP2 was downregulated in PCa cells and inhibited PCa proliferation and metastasis via the PI3K/AKT signaling pathway in vitro. In conclusion, LTBP2 was a promising diagnostic biomarker of BCR of PCa and had an important role in CD4+ T-cell recruitment. Moreover, it was associated with immunotherapy in patients with PCa who developed BCR, and it inhibited PCa proliferation and metastasis via the PI3K/AKT signaling pathway in vitro.
Collapse
Affiliation(s)
- Xiaowen Zhang
- Department of Urology, Affiliated Zhongda Hospital of South‑East University, Nanjing, Jiangsu 210009, P.R. China
| | - Chuanjie Tian
- Department of Urology, Langxi County People's Hospital, Xuancheng, Anhui 242100, P.R. China
| | - Chuanjie Tian
- Department of Urology, Langxi County People's Hospital, Xuancheng, Anhui 242100, P.R. China
| | - Jianbin Cheng
- Department of Urology Surgery, Heqiao Hospital, Yixing, Jiangsu 214200, P.R. China
| | - Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of South‑East University, Nanjing, Jiangsu 210009, P.R. China
| | - Menglan Li
- NHC Contraceptives Adverse Reaction Surveillance Center, Jiangsu Health Development Research Center, Nanjing, Jiangsu 210036, P.R. China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of South‑East University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
47
|
Wang C, Liu S, Zhang X, Wang Y, Guan P, Bu F, Wang H, Wang D, Fan Y, Hou S, Qiu Z. SKA3 is a prognostic biomarker and associated with immune infiltration in bladder cancer. Hereditas 2022; 159:20. [PMID: 35546682 PMCID: PMC9092687 DOI: 10.1186/s41065-022-00234-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Spindle and kinetochore‑associated complex subunit 3 (SKA3) has recently been considered a key regulator of carcinogenesis. However, the connection between SKA3 and immune cell infiltration remains unknown. METHODS The current study investigated the expression mode, prognostic effect, and functional role of SKA3 in different tumors, particularly bladder cancer using numerous databases, comprising TIMER, GEPIA, HPA, UALCAN, PrognoScan, and Kaplan-Meier Plotter. Differentially expressed gene and enrichment analyses were implemented on SKA3 using R packages "edgR" and "clusterProfiler". Immunohistochemistry was further used to validate the expression of SKA3 gene in bladder cancer. Following that, the relevance of SKA3 expression to immune infiltration level in bladder cancer was evaluated using TIMER. RESULTS Overall, the level of SKA3 expression in tumor tissue significantly increased than in normal tissue. In bladder cancer and other tumors, patients with high SKA3 expression levels had worse overall survival (OS) (p = 0.016), disease-specific survival (DSS) (p = 0.00004), and disease-free survival (DFS) (p = 0.032). Additionally, the major molecular functions for SKA3 included nuclear division, mitotic nuclear division, mitotic sister chromatid segregation, humoral immune response, and cell chemotaxis. Additionally, SKA3 expression was found to be positively associated with enhanced M2 macrophage and T helper (Th) 2 cell infiltration in bladder cancer. CONCLUSIONS Our study implies that SKA3 contributes to M2 macrophage and Th2 cell polarization by acting as an oncogene in bladder cancer. SKA3 might be a novel biomarker for evaluating prognosis and immune infiltration in bladder cancer.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Shasha Liu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Xinhong Zhang
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Yan Wang
- Department of Anesthesiology and Surgery, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Peng Guan
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Fanyou Bu
- Department of Traditional Chinese Medicine, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Hao Wang
- Department of Oncology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Dawen Wang
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Yi Fan
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Sichuan Hou
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China
| | - Zhilei Qiu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, 266071, Qingdao, Shandong, China.
| |
Collapse
|
48
|
Huang M, Dong W, Xie R, Wu J, Su Q, Li W, Yao K, Chen Y, Zhou Q, Zhang Q, Li W, Cheng L, Peng S, Chen S, Huang J, Chen X, Lin T. HSF1 facilitates the multistep process of lymphatic metastasis in bladder cancer via a novel PRMT5-WDR5-dependent transcriptional program. Cancer Commun (Lond) 2022; 42:447-470. [PMID: 35434944 PMCID: PMC9118058 DOI: 10.1002/cac2.12284] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/16/2022] [Accepted: 03/20/2022] [Indexed: 01/09/2023] Open
Abstract
Background Lymphatic metastasis has been associated with poor prognosis in bladder cancer patients with limited therapeutic options. Emerging evidence shows that heat shock factor 1 (HSF1) drives diversified transcriptome to promote tumor growth and serves as a promising therapeutic target. However, the roles of HSF1 in lymphatic metastasis remain largely unknown. Herein, we aimed to illustrate the clinical roles and mechanisms of HSF1 in the lymphatic metastasis of bladder cancer and explore its therapeutic potential. Methods We screened the most relevant gene to lymphatic metastasis among overexpressed heat shock factors (HSFs) and heat shock proteins (HSPs), and analyzed its clinical relevance in three cohorts. Functional in vitro and in vivo assays were performed in HSF1‐silenced and ‐regained models. We also used Co‐immunoprecipitation to identify the binding proteins of HSF1 and chromatin immunoprecipitation and dual‐luciferase reporter assays to investigate the transcriptional program directed by HSF1. The pharmacological inhibitor of HSF1, KRIBB11, was evaluated in popliteal lymph node metastasis models and patient‐derived xenograft models of bladder cancer. Results HSF1 expression was positively associated with lymphatic metastasis status, tumor stage, advanced grade, and poor prognosis of bladder cancer. Importantly, HSF1 enhanced the epithelial‐mesenchymal transition (EMT) of cancer cells in primary tumor to initiate metastasis, proliferation of cancer cells in lymph nodes, and macrophages infiltration to facilitate multistep lymphatic metastasis. Mechanistically, HSF1 interacted with protein arginine methyltransferase 5 (PRMT5) and jointly induced the monomethylation of histone H3 at arginine 2 (H3R2me1) and symmetric dimethylation of histone H3 at arginine 2 (H3R2me2s). This recruited the WD repeat domain 5 (WDR5)/mixed‐lineage leukemia (MLL) complex to increase the trimethylation of histone H3 at lysine 4 (H3K4me3); resulting in upregulation of lymphoid enhancer‐binding factor 1 (LEF1), matrix metallopeptidase 9 (MMP9), C‐C motif chemokine ligand 20 (CCL20), and E2F transcription factor 2 (E2F2). Application of KRIBB11 significantly inhibited the lymphatic metastasis of bladder cancer with no significant toxicity. Conclusion Our findings reveal a novel transcriptional program directed by the HSF1‐PRMT5‐WDR5 axis during the multistep process of lymphatic metastasis in bladder cancer. Targeting HSF1 could be a multipotent and promising therapeutic strategy for bladder cancer patients with lymphatic metastasis.
Collapse
Affiliation(s)
- Ming Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Wen Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, P. R. China
| | - Ruihui Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Jilin Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Wuguo Li
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Kai Yao
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Yuelong Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Qianghua Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Qiang Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Wenwen Li
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, P. R. China
| | - Liang Cheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Shengmeng Peng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Siting Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, P. R. China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, P. R. China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, P. R. China.,Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, P. R. China
| |
Collapse
|
49
|
Siladi AJ, Wang J, Florian AC, Thomas LR, Creighton JH, Matlock BK, Flaherty DK, Lorey SL, Howard GC, Fesik SW, Weissmiller AM, Liu Q, Tansey WP. WIN site inhibition disrupts a subset of WDR5 function. Sci Rep 2022; 12:1848. [PMID: 35115608 PMCID: PMC8813994 DOI: 10.1038/s41598-022-05947-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/19/2022] [Indexed: 11/09/2022] Open
Abstract
WDR5 nucleates the assembly of histone-modifying complexes and acts outside this context in a range of chromatin-centric processes. WDR5 is also a prominent target for pharmacological inhibition in cancer. Small-molecule degraders of WDR5 have been described, but most drug discovery efforts center on blocking the WIN site of WDR5, an arginine binding cavity that engages MLL/SET enzymes that deposit histone H3 lysine 4 methylation (H3K4me). Therapeutic application of WIN site inhibitors is complicated by the disparate functions of WDR5, but is generally guided by two assumptions-that WIN site inhibitors disable all functions of WDR5, and that changes in H3K4me drive the transcriptional response of cancer cells to WIN site blockade. Here, we test these assumptions by comparing the impact of WIN site inhibition versus WDR5 degradation on H3K4me and transcriptional processes. We show that WIN site inhibition disables only a specific subset of WDR5 activity, and that H3K4me changes induced by WDR5 depletion do not explain accompanying transcriptional responses. These data recast WIN site inhibitors as selective loss-of-function agents, contradict H3K4me as a relevant mechanism of action for WDR5 inhibitors, and indicate distinct clinical applications of WIN site inhibitors and WDR5 degraders.
Collapse
Affiliation(s)
- Andrew J Siladi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
| | - Jing Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrea C Florian
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
| | - Lance R Thomas
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
- Oncocyte Corporation, 2 International Drive, Suite 510, Nashville, TN, 37217, USA
| | - Joy H Creighton
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, 32132, USA
| | - Brittany K Matlock
- Vanderbilt University Medical Center Flow Cytometry Shared Resource, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - David K Flaherty
- Vanderbilt University Medical Center Flow Cytometry Shared Resource, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Shelly L Lorey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
| | - Gregory C Howard
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
| | - Stephen W Fesik
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - April M Weissmiller
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, 32132, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - William P Tansey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 465 21st Avenue South, Nashville, TN, 37232, USA.
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| |
Collapse
|
50
|
Miyake M, Nishimura N, Shimizu T, Ohnishi M, Kuwada M, Itami Y, Inoue T, Ohnishi K, Matsumoto Y, Yoshida T, Tatsumi Y, Shinohara M, Hori S, Morizawa Y, Gotoh D, Nakai Y, Anai S, Torimoto K, Aoki K, Fujii T, Tanaka N, Fujimoto K. Significant Improvement of Prognosis After the Advent of Immune Checkpoint Inhibitors in Patients with Advanced, Unresectable, or Metastatic Urothelial Carcinoma: A Propensity Score Matching and Inverse Probability of Treatment Weighting Analysis on Real-World Data. Cancer Manag Res 2022; 14:623-635. [PMID: 35210859 PMCID: PMC8858764 DOI: 10.2147/cmar.s348899] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/30/2022] [Indexed: 12/21/2022] Open
Abstract
Purpose The treatment landscape for advanced, unresectable, or metastatic urothelial carcinoma (aUC) has shifted substantially since the advent of immune checkpoint inhibitors (ICIs). We investigated the extent to which pembrolizumab therapy is superior to conventional chemotherapy as a second-line treatment. Patients and Methods A multicenter-derived database registered 454 patients diagnosed with aUC between 2008 and 2020. Of these, 94 patients (21%) who received second-line pembrolizumab and 75 (17%) who received second-line chemotherapy but never received third-line or later ICI therapy were included. We compared overall survival (OS) from the initial date of first-line chemotherapy between two groups by adjusting for prognostic factors through propensity score matching (PSM) and inverse probability of treatment weighting (IPTW). The IPTW-adjusted hazard ratio and 95% confidence interval were estimated using a multivariate Cox regression analysis. To identify patients who were more likely to benefit from second-line pembrolizumab than from chemotherapy, we performed a subgroup analysis for OS with an IPTW-adjusted model. Results The PSM-adjusted comparison showed a significant improvement in the prognosis with second-line pembrolizumab use (P = 0.01). The OS benefit with the advent of pembrolizumab was 8 months (18 months vs 26 months). Multivariable analyses using IPTW adjustment demonstrated that lymph node metastasis (P = 0.001), lung metastasis (P = 0.013), and bone metastasis (P = 0.003) were poor independent prognostic factors, and pembrolizumab use (P = 0.021) was a favorable independent prognostic factor. Subgroup analyses revealed that pembrolizumab was associated with survival benefits over chemotherapy in all subgroups, including young patients (age <70 years), those who received radical surgery, and those without visceral metastasis. Conclusion We demonstrated a significant improvement in prognosis after the advent of pembrolizumab for patients with aUC. ICIs should not be restricted based on patient characteristics.
Collapse
Affiliation(s)
- Makito Miyake
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
- Correspondence: Makito Miyake, Department of Urology, Nara Medical University, 840 Shijo-cho, Nara, 634-8522, Japan, Tel +81-744-22-3051 (ext 2338), Fax +81-744-22-9282, Email
| | - Nobutaka Nishimura
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
- Department of Urology, Okanami General Hospital, Iga, Mie, Japan
| | - Takuto Shimizu
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
- Department of Urology, Saiseikai Chuwa Hospital, Nara, Japan
| | - Mikiko Ohnishi
- Department of Urology, Matsusaka Chuo General Hospital, Matsusaka, Mie, Japan
| | - Masaomi Kuwada
- Department of Urology, Matsusaka Chuo General Hospital, Matsusaka, Mie, Japan
| | - Yoshitaka Itami
- Department of Urology, Nara Prefecture General Medical Center, Nara, Japan
| | - Takeshi Inoue
- Department of Urology, Nara Prefecture General Medical Center, Nara, Japan
| | - Kenta Ohnishi
- Department of Urology, Hoshigaoka Medical Center, Hirakata, Osaka, Japan
| | | | | | - Yoshihiro Tatsumi
- Department of Urology, JCHO Yamato Koriyama Hospital, Koriyama, Nara, Japan
| | | | - Shunta Hori
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Yosuke Morizawa
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Daisuke Gotoh
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Yasushi Nakai
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Satoshi Anai
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Kazumasa Torimoto
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Katsuya Aoki
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Tomomi Fujii
- Department of Diagnostic Pathology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Nobumichi Tanaka
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
- Department of Prostate Brachytherapy, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| | - Kiyohide Fujimoto
- Department of Urology, Nara Medical University, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|