1
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
2
|
Shao X, Yu L, Li C, Qian J, Yang X, Yang H, Liao J, Fan X, Xu X, Fan X. Extracellular vesicle-derived miRNA-mediated cell-cell communication inference for single-cell transcriptomic data with miRTalk. Genome Biol 2025; 26:95. [PMID: 40229908 PMCID: PMC11998287 DOI: 10.1186/s13059-025-03566-x] [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: 06/26/2023] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
MicroRNAs are released from cells in extracellular vesicles (EVs), representing an essential mode of cell-cell communication (CCC) via a regulatory effect on gene expression. Single-cell RNA-sequencing technologies have ushered in an era of elucidating CCC at single-cell resolution. Herein, we present miRTalk, a pioneering approach for inferring CCC mediated by EV-derived miRNA-target interactions (MiTIs). The benchmarking against simulated and real-world datasets demonstrates the superior performance of miRTalk, and the application to four disease scenarios reveals the in-depth MiTI-mediated CCC mechanisms. Collectively, miRTalk can infer EV-derived MiTI-mediated CCC with scRNA-seq data, providing new insights into the intercellular dynamics of biological processes.
Collapse
Affiliation(s)
- Xin Shao
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women'S Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- The Joint-Laboratory of Clinical Multi-Omics Research Between, Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo, 315012, China.
| | - Lingqi Yu
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women'S Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chengyu Li
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jingyang Qian
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinyu Yang
- The Center for Integrated Oncology and Precision Medicine, School of Medicine, Affiliated Hangzhou First People'S Hospital, Westlake University, Hangzhou, 310006, China
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Haihong Yang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310027, China
| | - Jie Liao
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xueru Fan
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao Xu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People'S Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310024, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
| | - Xiaohui Fan
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women'S Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- The Joint-Laboratory of Clinical Multi-Omics Research Between, Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo, 315012, China.
| |
Collapse
|
3
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [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: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
4
|
Li B, Gan J, Li T, Chen J, Kuang Y, Li J, Yin H. Comprehensive analysis of RNA methylation-related genes to identify molecular cluster for predicting prognosis and immune profiles in bladder cancer. Sci Rep 2025; 15:9147. [PMID: 40097551 PMCID: PMC11914693 DOI: 10.1038/s41598-025-93674-2] [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: 05/28/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
m6A, m5C and m7G are common types of RNA methylation modifications that are widely involved in key mechanisms regulating malignancy. However, the role of RNA methylation-related genes in the immune microenvironment of bladder cancer (BLCA) remains elusive. In this study, we established RNA methylation molecular subtypes by analyzing the TCGA and GEO datasets. Risk model and nomogram were constructed by LASSO and multivariate Cox regression analysis and validated by external datasets. Genetic variations, functional enrichment analysis and immune cell infiltration were analyzed. The expression levels of hub genes were detected by real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). The effect of FN1 on cellular function was determined using experimental assays. Finally, we identified a 7-gene signature associated with BLCA prognosis. GSE19423 validated the predictive value of the risk model. The IMvigor210 data showed the model had promising predictive efficacy for BLCA immunotherapy. Significant differences in biological function, immune cell infiltration and drug sensitivity were observed between high- and low-risk groups. Furthermore, FN1 was upregulated in BLCA, as determined by qRT-PCR and IHC. Depletion of FN1 using siRNA impaired cell motility in T24 and 5637 cells. In conclusion, RNA methylation-related risk model can predict the prognosis, immune landscape and response to immunotherapy in BLCA. Among the 7-gene signature, FN1 is a pivotal gene that promotes the migration of bladder cancer cells.
Collapse
Affiliation(s)
- Bo Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junlin Gan
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tinghao Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junrui Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Central Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Youlin Kuang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jie Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Hubin Yin
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
5
|
Song B, Wu P, Wan C, Sun Q, Kong G. Integrating single cell and bulk RNA sequencing data identifies RBM17 as a novel response biomarker for immunotherapy in bladder cancer. Virchows Arch 2024; 485:1133-1150. [PMID: 39453457 DOI: 10.1007/s00428-024-03952-z] [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: 06/11/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
Checkpoint inhibitors (CPIs) have been widely applied in the treatment of patients with bladder cancer (BLCA). However, there is still unmet need to dissect response predict biomarkers. To uncover CPI response-related marker genes in cancer cells, we utilized SCISSOR, integrating single-cell RNA and bulk RNA sequencing data. Transcriptomic and clinical data from IMvigor210, UNC-108, and BCAN/HCRN datasets were collected to evaluate and validate the identified biomarkers and signatures. Additionally, we analyzed TCGA-BLCA and local-BLCA RNA-seq data to investigate alternative splicing events (ASEs). Cell viability was assessed in T24 and UMUC3 cells with RBM17 upregulation or downregulation. Through SCISSOR analysis, we discovered that the expression levels of RBM17, TAP1, and PSMB8 were significantly associated with CPI response. Since PSMB8 displayed a highly positive correlation with TAP1, we developed a CPI response score (CRS) signature based on the expression profiles of RBM17 and TAP1. The CRS demonstrated robust predictive capacity in IMvigor210, UNC-108, and BCAN/HCRN datasets and was associated with higher tumor mutational burden (TMB), PD-L1 expression, and unique genomic features. Notably, RBM17 was not linked to the clinical outcomes of BLCA patients but positively correlated with BLCA cell proliferation in vitro. In the meantime, RBM17 was correlated with higher activity in core biological pathways, including antigen processing machinery, CD8 + T effector cells, cell cycle, DNA damage repair, epithelial-mesenchymal transition, histone regulation, and immune checkpoints. Moreover, the high-RBM17 group showed enrichment of LumU/Ba/sq subtypes but fewer FGFR3 alterations. Lastly, RBM17 significantly upregulated ASEs in BLCA samples, leading to higher neoantigen levels, a more inflamed tumor microenvironment, and improved CPI response. RBM17 is associated with higher ASEs and neoantigen levels, thereby potentiating the efficacy of CPI in BLCA. The established predictive signature, utilizing only two genes, has the potential to streamline clinical applications, providing a cost-effective alternative to expensive genomic, transcriptomic, and biological feature tests.
Collapse
Affiliation(s)
- Bo Song
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China
| | - Peishan Wu
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China.
| | - Chong Wan
- Precision Medicine Center, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, 314001, Zhejiang, China
| | - Qiangqiang Sun
- Department of Precision Medicine, Accb Co. Ltd., Jiaxing, 314001, China
| | - Guangqi Kong
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Road, Tongzhou District, Beijing, 101149, China
| |
Collapse
|
6
|
Zeng L, Zhang L, Li L, Liao X, Yin C, Zhang L, Chen X, Sun J. RNA sequencing identifies lung cancer lineage and facilitates drug repositioning. PeerJ 2024; 12:e18159. [PMID: 39346064 PMCID: PMC11430167 DOI: 10.7717/peerj.18159] [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: 05/27/2024] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Recent breakthrough therapies have improved survival rates in non-small cell lung cancer (NSCLC), but a paradigm for prospective confirmation is still lacking. Patientdatasets were mainly downloaded from TCGA, CPTAC and GEO. We conducted downstream analysis by collecting metagenes and generated 42-gene subtype classifiers to elucidate biological pathways. Subsequently, scRNA, eRNA, methylation, mutation, and copy number variation were depicted from a phenotype perspective. Enhancing the clinical translatability of molecular subtypes, preclinical models including CMAP, CCLE, and GDSC were utilized for drug repositioning. Importantly, we verified the presence of previously described three phenotypes including bronchioid, neuroendocrine, and squamoid. Poor prognosis was seen in squamoid and neuroendocrine clusters for treatment-naive and immunotherapy populations. The neuroendocrine cluster was dominated by STK11 mutations and 14q13.3 amplifications, whose related methylated loci are predictive of immunotherapy. And the greatest therapeutic potential lies in the bronchioid cluster. We further estimated the relative cell abundance of the tumor microenvironment (TME), specific cell types could be reflected among three clusters. Meanwhile, the higher portion of immune cell infiltration belonged to bronchioid and squamoid, not the neuroendocrine cluster. In drug repositioning, MEK inhibitors resisted bronchioid but were squamoid-sensitive. To conceptually validate compounds/targets, we employed RNA-seq and CCK-8/western blot assays. Our results indicated that dinaciclib and alvocidib exhibited similar activity and sensitivity in the neuroendocrine cluster. Also, a lineage factor named KLF5 recognized by inferred transcriptional factors activity could be suppressed by verteporfin.
Collapse
Affiliation(s)
- Longjin Zeng
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Longyao Zhang
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| | - Lingchen Li
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| | - Xingyun Liao
- Affiliated Tumor Hospital, Department of Oncology, Chongqing, China
| | - Chenrui Yin
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| | - Lincheng Zhang
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiewan Chen
- Department of Basic Medicine, Army Medical University, Chongqing, China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Chongqing, China
| |
Collapse
|
7
|
Yan Z, Liu Y, Wang M, Wang L, Chen Z, Liu X. A novel signature constructed by mitochondrial function and cell death-related gene for the prediction of prognosis in bladder cancer. Sci Rep 2024; 14:14667. [PMID: 38918587 PMCID: PMC11199696 DOI: 10.1038/s41598-024-65594-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024] Open
Abstract
Bladder urothelial carcinoma (BLCA) presents a persistent challenge in clinical management. Despite recent advancements demonstrating the BLCA efficacy of immune checkpoint inhibitors (ICI) in BLCA patients, there remains a critical need to identify and expand the subset of individuals who benefit from this treatment. Mitochondria, as pivotal regulators of various cell death pathways in eukaryotic cells, exert significant influence over tumor cell fate and survival. In this study, our objective was to investigate biomarkers centered around mitochondrial function and cell death mechanisms to facilitate prognostic prediction and guide therapeutic decision-making in BLCA. Utilizing ssGSEA and LASSO regression, we developed a prognostic signature termed mitochondrial function and cell death (mtPCD). Subsequently, we evaluated the associations between mtPCD score and diverse clinical outcomes, including prognosis, functional pathway enrichment, immune cell infiltration, immunotherapy response analysis and drug sensitivity, within high- and low-risk subgroups. Additionally, we employed single-cell level functional assays, RT-qPCR, and immunohistochemistry to validate the differential expression of genes comprising the mtPCD signature. The mtPCD signature comprises a panel of 10 highly influential genes, strongly correlated with survival outcomes in BLCA patients and exhibiting robust predictive capabilities. Importantly, individuals classified as high-risk according to mtPCD score displayed a subdued overall immune response, characterized by diminished immunotherapeutic efficacy. In summary, our findings highlight the development of a novel prognostic signature, which not only holds promise as a biomarker for BLCA prognosis but also offers insights into the immune landscape of BLCA. This paradigm may pave the way for personalized treatment strategies in BLCA management.
Collapse
Affiliation(s)
- Zhiwei Yan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yunxun Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Minghui Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
8
|
Wang L, Izadmehr S, Sfakianos JP, Tran M, Beaumont KG, Brody R, Cordon-Cardo C, Horowitz A, Sebra R, Oh WK, Bhardwaj N, Galsky MD, Zhu J. Single-cell transcriptomic-informed deconvolution of bulk data identifies immune checkpoint blockade resistance in urothelial cancer. iScience 2024; 27:109928. [PMID: 38812546 PMCID: PMC11133924 DOI: 10.1016/j.isci.2024.109928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/23/2023] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Interactions within the tumor microenvironment (TME) significantly influence tumor progression and treatment responses. While single-cell RNA sequencing (scRNA-seq) and spatial genomics facilitate TME exploration, many clinical cohorts are assessed at the bulk tissue level. Integrating scRNA-seq and bulk tissue RNA-seq data through computational deconvolution is essential for obtaining clinically relevant insights. Our method, ProM, enables the examination of major and minor cell types. Through evaluation against existing methods using paired single-cell and bulk RNA sequencing of human urothelial cancer (UC) samples, ProM demonstrates superiority. Application to UC cohorts treated with immune checkpoint inhibitors reveals pre-treatment cellular features associated with poor outcomes, such as elevated SPP1 expression in macrophage/monocytes (MM). Our deconvolution method and paired single-cell and bulk tissue RNA-seq dataset contribute novel insights into TME heterogeneity and resistance to immune checkpoint blockade.
Collapse
Affiliation(s)
- Li Wang
- Department of Precision Medicine, Aitia, Somerville, MA 02143, USA
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
| | - Sudeh Izadmehr
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
| | - John P. Sfakianos
- Department of Urology; Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michelle Tran
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rachel Brody
- Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carlos Cordon-Cardo
- Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amir Horowitz
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William K. Oh
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
| | - Nina Bhardwaj
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Matthew D. Galsky
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
| | - Jun Zhu
- Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY 10029, USA
| |
Collapse
|
9
|
Huang J, Du Y, Stucky A, Kelly KR, Zhong JF, Sun F. DeepDecon accurately estimates cancer cell fractions in bulk RNA-seq data. PATTERNS (NEW YORK, N.Y.) 2024; 5:100969. [PMID: 38800361 PMCID: PMC11117059 DOI: 10.1016/j.patter.2024.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/15/2024] [Accepted: 03/21/2024] [Indexed: 05/29/2024]
Abstract
Understanding the cellular composition of a disease-related tissue is important in disease diagnosis, prognosis, and downstream treatment. Recent advances in single-cell RNA-sequencing (scRNA-seq) technique have allowed the measurement of gene expression profiles for individual cells. However, scRNA-seq is still too expensive to be used for large-scale population studies, and bulk RNA-seq is still widely used in such situations. An essential challenge is to deconvolve cellular composition for bulk RNA-seq data based on scRNA-seq data. Here, we present DeepDecon, a deep neural network model that leverages single-cell gene expression information to accurately predict the fraction of cancer cells in bulk tissues. It provides a refining strategy in which the cancer cell fraction is iteratively estimated by a set of trained models. When applied to simulated and real cancer data, DeepDecon exhibits superior performance compared to existing decomposition methods in terms of accuracy.
Collapse
Affiliation(s)
- Jiawei Huang
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuxuan Du
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andres Stucky
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Kevin R. Kelly
- Division of Hematology, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiang F. Zhong
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Fengzhu Sun
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
10
|
Ma Y, Jin J, Xue Z, Zhao J, Cai W, Zhang W. Integrated multi-omics analysis and machine learning developed a prognostic model based on mitochondrial function in a large multicenter cohort for Gastric Cancer. J Transl Med 2024; 22:381. [PMID: 38654380 PMCID: PMC11040813 DOI: 10.1186/s12967-024-05109-7] [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: 01/10/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common and aggressive type of cancer worldwide. Despite recent advancements in its treatment, the prognosis for patients with GC remains poor. Understanding the mechanisms of cell death in GC, particularly those related to mitochondrial function, is crucial for its development and progression. However, more research is needed to investigate the significance of the interaction between mitochondrial function and GC cell death. METHODS We employed a robust computational framework to investigate the role of mitochondria-associated proteins in the progression of GC in a cohort of 1,199 GC patients. Ten machine learning algorithms were utilized and combined into 101 unique combinations. Ultimately, we developed a Mitochondrial-related-Score (MitoScore) using the machine learning model that exhibited the best performance. We observed the upregulation of LEMT2 and further explored its function in tumor progression. Mitochondrial functions were assessed by measuring mitochondrial ATP, mitochondrial membrane potential, and levels of lactate, pyruvate, and glucose. RESULTS MitoScore showed significant correlations with GC immune and metabolic functions. The higher MitoScore subgroup exhibited enriched metabolic pathways and higher immune activity. Overexpression of LETM2 (leucine zipper and EF-hand containing transmembrane protein 2) significantly enhanced tumor proliferation and metastasis. LETM2 plays a role in promoting GC cell proliferation by activating the mTOR pathway, maintaining mitochondrial homeostasis, and promoting glycolysis. CONCLUSION The powerful machine learning framework highlights the significant potential of MitoScore in providing valuable insights and accurate assessments for individuals with GC. This study also enhances our understanding of LETM2 as an oncogene signature in GC. LETM2 may promote tumor progression by maintaining mitochondrial health and activating glycolysis, offering potential targets for diagnosis, treatment, and prognosis of GC.
Collapse
Affiliation(s)
- Yimeng Ma
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Jin
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Wenzhou Medical University, Wenzhou, China
| | - Zixuan Xue
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jungang Zhao
- Department of Urology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Weiyang Cai
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wanli Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
11
|
Chatterjee D, Mou SI, Sultana T, Hosen MI, Faruk MO. Identification and validation of prognostic signature genes of bladder cancer by integrating methylation and transcriptomic analysis. Sci Rep 2024; 14:368. [PMID: 38172584 PMCID: PMC10764961 DOI: 10.1038/s41598-023-50740-x] [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: 11/05/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024] Open
Abstract
Being a frequent malignant tumor of the genitourinary system, Bladder Urothelial Carcinoma (BLCA) has a poor prognosis. This study focused on identifying and validating prognostic biomarkers utilizing methylation, transcriptomics, and clinical data from The Cancer Genome Atlas Bladder Urothelial Carcinoma (TCGA BLCA) cohort. The impact of altered differentially methylated hallmark pathway genes was subjected to clustering analysis to observe changes in the transcriptional landscape on BLCA patients and identify two subtypes of patients from the TCGA BLCA population where Subtype 2 was associated with the worst prognosis with a p-value of 0.00032. Differential expression and enrichment analysis showed that subtype 2 was enriched in immune-responsive and cancer-progressive pathways, whereas subtype 1 was enriched in biosynthetic pathways. Following, regression and network analyses revealed Epidermal Growth Factor Receptor (EGFR), Fos-related antigen 1 (FOSL1), Nuclear Factor Erythroid 2 (NFE2), ADP-ribosylation factor-like protein 4D (ARL4D), SH3 domain containing ring finger 2 (SH3RF2), and Cadherin 3 (CDH3) genes to be the most significant prognostic gene markers. These genes were used to construct a risk model that separated the BLCA patients into high and low-risk groups. The risk model was also validated in an external dataset by performing survival analysis between high and low-risk groups with a p-value < 0.001 and the result showed the high group was significantly associated with poor prognosis compared to the low group. Single-cell analyses revealed the elevated level of these genes in the tumor microenvironment and associated with immune response. High-grade patients also tend to have a high expression of these genes compared to low-grade patients. In conclusion, this research developed a six-gene signature that is pertinent to the prediction of overall survival (OS) and might contribute to the advancement of precision medicine in the management of bladder cancer.
Collapse
Affiliation(s)
- Dipankor Chatterjee
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Sadia Islam Mou
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Tamanna Sultana
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Md Ismail Hosen
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Md Omar Faruk
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
12
|
Tuo Z, Feng D, Jiang Z, Bi L, Yang C, Wang Q. Unveiling clinical significance and tumor immune landscape of CXCL12 in bladder cancer: Insights from multiple omics analysis. Chin J Cancer Res 2023; 35:686-701. [PMID: 38204439 PMCID: PMC10774138 DOI: 10.21147/j.issn.1000-9604.2023.06.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024] Open
Abstract
Objective The interplay between chemokine C-X-C motif ligand 12 (CXCL12) and its specific receptors is known to trigger various signaling pathways, contributing to tumor proliferation and metastasis. Consequently, targeting this signaling axis has emerged as a potential strategy in cancer therapy. However, the precise role of CXCL12 in clinical therapy, especially in immunotherapy for bladder cancer (BCa), remains poorly elucidated. Methods We gathered multiple omics data from public databases to unveil the clinical relevance and tumor immune landscape associated with CXCL12 in BCa patients. Univariate and multivariate Cox regression analyses were employed to assess the independent prognostic significance of CXCL12 expression and formulate a nomogram. The expression of CXCL12 in BCa cell lines and clinical tissue samples was validated using enzyme-linked immunosorbent assays (ELISA) and immunohistochemistry (IHC). Results While transcriptional expression of CXCL12 exhibited a decrease in nearly all tumor tissues, CXCL12 methylation expression was notably increased in BCa tissues. Single-cell RNA analysis highlighted tissue stem cells and endothelial cells as the primary sources expressing CXCL12. Abnormal CXCL12 expression, based on transcriptional and methylation levels, correlated with various clinical characteristics in BCa patients. Functional analysis indicated enrichment of CXCL12 and its co-expression genes in immune regulation and cell adhesion. The immune landscape analysis unveiled a significant association between CXCL12 expression and M2 macrophages (CD163+ cells) in BCa tissues. Notably, CXCL12 expression emerged as a potential predictor of immunotherapy response and chemotherapy drug sensitivity in BCa patients. Conclusions Taken together, these findings suggest aberrant production of CXCL12 in BCa tissues, potentially influencing the treatment responses of affected individuals.
Collapse
Affiliation(s)
- Zhouting Tuo
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Rehabilitation, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhiwei Jiang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Liangkuan Bi
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Chao Yang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Qi Wang
- Department of Urology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
13
|
Maden SK, Kwon SH, Huuki-Myers LA, Collado-Torres L, Hicks SC, Maynard KR. Challenges and opportunities to computationally deconvolve heterogeneous tissue with varying cell sizes using single-cell RNA-sequencing datasets. Genome Biol 2023; 24:288. [PMID: 38098055 PMCID: PMC10722720 DOI: 10.1186/s13059-023-03123-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Deconvolution of cell mixtures in "bulk" transcriptomic samples from homogenate human tissue is important for understanding disease pathologies. However, several experimental and computational challenges impede transcriptomics-based deconvolution approaches using single-cell/nucleus RNA-seq reference atlases. Cells from the brain and blood have substantially different sizes, total mRNA, and transcriptional activities, and existing approaches may quantify total mRNA instead of cell type proportions. Further, standards are lacking for the use of cell reference atlases and integrative analyses of single-cell and spatial transcriptomics data. We discuss how to approach these key challenges with orthogonal "gold standard" datasets for evaluating deconvolution methods.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA.
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Song Z, Su M, Li X, Xie J, Han F, Yao J. A novel endoplasmic reticulum stress-related lncRNA signature for prognosis prediction and immune response evaluation in Stomach adenocarcinoma. BMC Gastroenterol 2023; 23:432. [PMID: 38066437 PMCID: PMC10709857 DOI: 10.1186/s12876-023-03001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 10/16/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is a significant contributor to cancer-related mortality worldwide. Although previous research has identified endoplasmic reticulum stress (ERS) as a regulator of various tumor-promoting properties of cancer cells, the impact of ERS-related long non-coding RNAs (lncRNAs) on STAD prognosis has not yet been investigated. Therefore, our study aims to develop and validate an ERS-related lncRNA signature that can accurately predict the prognosis of STAD patients. METHODS We collected RNA expression profiles and clinical data of STAD patients from The Cancer Genome Atlas (TCGA) and identified ERS-related genes from the Molecular Signature Database (MSigDB). Co-expression analysis enabled us to identify ERS-related lncRNAs, and we applied univariate Cox, least absolute shrinkage, and selection operator (LASSO), and multivariate Cox regression analyses to construct a predictive signature comprising of 9 ERS-related lncRNAs. We assessed the prognostic accuracy of our signature using Kaplan-Meier survival analysis, and validated our predictive signature in an independent gene expression omnibus (GEO) cohort. We also performed tumor mutational burden (TMB) and tumor immune microenvironment (TIME) analyses. Enrichment analysis was used to investigate the functions and biological processes of the signature, and we identified two distinct STAD patient subgroups through consensus clustering. Finally, we performed drug sensitivity analysis and immunologic efficacy analysis to explore further insights. RESULTS The 9 ERS related-lncRNAs signature demonstrated satisfactory predictive performance as an independent prognostic marker and was significantly associated with STAD clinicopathological characteristics. Furthermore, patients in the high-risk group displayed a worse STAD prognosis than those in the low-risk group. Notably, gene set enrichment analysis (GSEA) revealed significant enrichment of extracellular matrix pathways in the high-risk group, indicating their involvement in STAD progression. Additionally, the high-risk group exhibited significantly lower TMB expression levels than the low-risk group. Consensus clustering revealed two distinct STAD patient subgroups, with Cluster 1 exhibiting higher immune cell infiltration and more active immune functions. Drug sensitivity analysis suggested that the low-risk group was more responsive to oxaliplatin, epirubicinl, and other drugs. CONCLUSION Our study highlights the crucial regulatory roles of ERS-related lncRNAs in STAD, with significant clinical implications. The 9-lncRNA signature we have constructed represents a reliable prognostic indicator that has the potential to inform more personalized treatment decisions for STAD patients. These findings shed new light on the pathogenesis of STAD and its underlying molecular mechanisms, offering opportunities for novel therapeutic strategies to be developed for STAD patients.
Collapse
Affiliation(s)
- Zhaoxiang Song
- Depratment of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengge Su
- Depratment of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiangyu Li
- Depratment of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinlin Xie
- Depratment of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fei Han
- Depratment of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianning Yao
- Depratment of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
15
|
Zhang S, Li X, Zheng Y, Liu J, Hu H, Zhang S, Kuang W. Single cell and bulk transcriptome analysis identified oxidative stress response-related features of Hepatocellular Carcinoma. Front Cell Dev Biol 2023; 11:1191074. [PMID: 37842089 PMCID: PMC10568628 DOI: 10.3389/fcell.2023.1191074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Background: Hepatocellular Carcinoma (HCC) is a common lethal digestive system tumor. The oxidative stress mechanism is crucial in the HCC genesis and progression. Methods: Our study analyzed single-cell and bulk sequencing data to compare the microenvironment of non-tumor liver tissues and HCC tissues. Through these analyses, we aimed to investigate the effect of oxidative stress on cells in the HCC microenvironment and identify critical oxidative stress response-related genes that impact the survival of HCC patients. Results: Our results showed increased oxidative stress in HCC tissue compared to non-tumor tissue. Immune cells in the HCC microenvironment exhibited higher oxidative detoxification capacity, and oxidative stress-induced cell death of dendritic cells was attenuated. HCC cells demonstrated enhanced communication with immune cells through the MIF pathway in a highly oxidative hepatoma microenvironment. Meanwhile, using machine learning and Cox regression screening, we identified PRDX1 as a predictor of early occurrence and prognosis in patients with HCC. The expression level of PRDX1 in HCC was related to dysregulated ribosome biogenesis and positively correlated with the expression of immunological checkpoints (PDCD1LG2, CTLA4, TIGIT, LAIR1). High PRDX1 expression in HCC patients correlated with better sensitivity to immunotherapy agents such as sorafenib, IGF-1R inhibitor, and JAK inhibitor. Conclusion: In conclusion, our study unveiled variations in oxidative stress levels between non-tumor liver and HCC tissues. And we identified oxidative stress gene markers associated with hepatocarcinogenesis development, offering novel insights into the oxidative stress response mechanism in HCC.
Collapse
Affiliation(s)
- Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinyu Li
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yilu Zheng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiahui Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Hu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Dongguan Key Laboratory of Chronic Inflammatory Diseases, School of Pharmacy, The First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
16
|
Qin Y, Zu X, Li Y, Han Y, Tan J, Cai C, Shen E, Liu P, Deng G, Feng Z, Wu W, Peng Y, Liu Y, Ma J, Zeng S, Chen Y, Shen H. A cancer-associated fibroblast subtypes-based signature enables the evaluation of immunotherapy response and prognosis in bladder cancer. iScience 2023; 26:107722. [PMID: 37694141 PMCID: PMC10485638 DOI: 10.1016/j.isci.2023.107722] [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: 11/28/2022] [Revised: 01/28/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023] Open
Abstract
Bladder cancer (BLCA) is one of the most prevalent and heterogeneous urinary malignant tumors. Previous researches have reported a significant association between cancer-associated fibroblasts (CAFs) and poor prognosis of tumor patients. However, uncertainty surrounds the role of CAFs in the BLCA tumor microenvironment, necessitating further investigation into the CAFs-related gene signatures in BLCA. In this study, we identified three CAF subtypes in BLCA according to single-cell RNA-seq data and constructed CAFs-related risk score (CRRS) by screening 102,714 signatures. The survival analysis, ROC curves, and nomogram suggested that CRRS was a valuable predictor in 2,042 patients from 9 available public datasets and Xiangya real-world cohort. We further revealed the significant correlation between CRRS and clinicopathological characteristics, genome alterations, and epithelial-mesenchymal transition (EMT). A high CRRS indicated a non-inflamed phenotype and a lower remission rate of immunotherapy in BLCA. In conclusion, the CRRS had the potential to predict the prognosis and immunotherapy response of BLCA patients.
Collapse
Affiliation(s)
- Yiming Qin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiongbing Zu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Edward Shen
- Department of Life Science, McMaster University, Hamilton L8S 4L8, ON, Canada
| | - Ping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ganlu Deng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530022, Guangxi, China
| | - Ziyang Feng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yinghui Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yongting Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jiayao Ma
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
17
|
Qin H, Abulaiti A, Maimaiti A, Abulaiti Z, Fan G, Aili Y, Ji W, Wang Z, Wang Y. Integrated machine learning survival framework develops a prognostic model based on inter-crosstalk definition of mitochondrial function and cell death patterns in a large multicenter cohort for lower-grade glioma. J Transl Med 2023; 21:588. [PMID: 37660060 PMCID: PMC10474752 DOI: 10.1186/s12967-023-04468-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/24/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Lower-grade glioma (LGG) is a highly heterogeneous disease that presents challenges in accurately predicting patient prognosis. Mitochondria play a central role in the energy metabolism of eukaryotic cells and can influence cell death mechanisms, which are critical in tumorigenesis and progression. However, the prognostic significance of the interplay between mitochondrial function and cell death in LGG requires further investigation. METHODS We employed a robust computational framework to investigate the relationship between mitochondrial function and 18 cell death patterns in a cohort of 1467 LGG patients from six multicenter cohorts worldwide. A total of 10 commonly used machine learning algorithms were collected and subsequently combined into 101 unique combinations. Ultimately, we devised the mitochondria-associated programmed cell death index (mtPCDI) using machine learning models that exhibited optimal performance. RESULTS The mtPCDI, generated by combining 18 highly influential genes, demonstrated strong predictive performance for prognosis in LGG patients. Biologically, mtPCDI exhibited a significant correlation with immune and metabolic signatures. The high mtPCDI group exhibited enriched metabolic pathways and a heightened immune activity profile. Of particular importance, our mtPCDI maintains its status as the most potent prognostic indicator even following adjustment for potential confounding factors, surpassing established clinical models in predictive strength. CONCLUSION Our utilization of a robust machine learning framework highlights the significant potential of mtPCDI in providing personalized risk assessment and tailored recommendations for metabolic and immunotherapy interventions for individuals diagnosed with LGG. Of particular significance, the signature features highly influential genes that present further prospects for future investigations into the role of PCD within mitochondrial function.
Collapse
Affiliation(s)
- Hu Qin
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Aimitaji Abulaiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Aierpati Maimaiti
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Zulihuma Abulaiti
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang, China
| | - Guofeng Fan
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Yirizhati Aili
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Wenyu Ji
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Zengliang Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China
| | - Yongxin Wang
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, No. 137, South Liyushan Road, Xinshi District, Urumqi City, 830054, Xinjiang, China.
| |
Collapse
|
18
|
Li H, Chen J, Li Z, Chen M, Ou Z, Mo M, Wang R, Tong S, Liu P, Cai Z, Zhang C, Liu Z, Deng D, Liu J, Cheng C, Hu J, Zu X. S100A5 Attenuates Efficiency of Anti-PD-L1/PD-1 Immunotherapy by Inhibiting CD8 + T Cell-Mediated Anti-Cancer Immunity in Bladder Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300110. [PMID: 37414584 PMCID: PMC10477882 DOI: 10.1002/advs.202300110] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/11/2023] [Indexed: 07/08/2023]
Abstract
Although immune checkpoint blockade (ICB) therapies have been approved for bladder cancer (BLCA), only a minority of patients respond to these therapies, and there is an urgent need to explore combined therapies. Systematic multi-omics analysis identified S100A5 as a novel immunosuppressive target for BLCA. The expression of S100A5 in malignant cells inhibited CD8+ T cell recruitment by decreasing pro-inflammatory chemokine secretion. Furthermore, S100A5 attenuated effector T cell killing of cancer cells by inhibiting CD8+ T cell proliferation and cytotoxicity. In addition, S100A5 acted as an oncogene, thereby promoting tumor proliferation and invasion. Targeting S100A5 synergized with the efficacy of anti-PD-1 treatment by enhancing infiltration and cytotoxicity of CD8+ T cells in vivo. Clinically, there was a spatially exclusive relationship between S100A5+ tumor cells and CD8+ T cells in tissue microarrays. Moreover, S100A5 negatively correlated with immunotherapy efficacy in our real-world and several public immunotherapy cohorts. In summary, S100A5 shapes a non-inflamed tumor microenvironment in BLCA by inhibiting the secretion of pro-inflammatory chemokines and the recruitment and cytotoxicity of CD8+ T cells. Targeting S100A5 converts cold tumors into hot tumors, thus enhancing the efficacy of ICB therapy in BLCA.
Collapse
Affiliation(s)
- Huihuang Li
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Jinbo Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Zhenghao Li
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research and Division of Hepato‐Biliary‐Pancreatic SurgeryDepartment of General SurgeryThe Second Xiangya HospitalCentral South UniversityChangsha410011China
| | - Minfeng Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Zhenyu Ou
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Miao Mo
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Ruizhe Wang
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Shiyu Tong
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Peihua Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Zhiyong Cai
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Chunyu Zhang
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Zhi Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Dingshan Deng
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Jinhui Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Chunliang Cheng
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Jiao Hu
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xiongbing Zu
- Department of UrologyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
19
|
Maden SK, Kwon SH, Huuki-Myers LA, Collado-Torres L, Hicks SC, Maynard KR. Challenges and opportunities to computationally deconvolve heterogeneous tissue with varying cell sizes using single cell RNA-sequencing datasets. ARXIV 2023:arXiv:2305.06501v1. [PMID: 37214135 PMCID: PMC10197733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Deconvolution of cell mixtures in "bulk" transcriptomic samples from homogenate human tissue is important for understanding the pathologies of diseases. However, several experimental and computational challenges remain in developing and implementing transcriptomics-based deconvolution approaches, especially those using a single cell/nuclei RNA-seq reference atlas, which are becoming rapidly available across many tissues. Notably, deconvolution algorithms are frequently developed using samples from tissues with similar cell sizes. However, brain tissue or immune cell populations have cell types with substantially different cell sizes, total mRNA expression, and transcriptional activity. When existing deconvolution approaches are applied to these tissues, these systematic differences in cell sizes and transcriptomic activity confound accurate cell proportion estimates and instead may quantify total mRNA content. Furthermore, there is a lack of standard reference atlases and computational approaches to facilitate integrative analyses, including not only bulk and single cell/nuclei RNA-seq data, but also new data modalities from spatial -omic or imaging approaches. New multi-assay datasets need to be collected with orthogonal data types generated from the same tissue block and the same individual, to serve as a "gold standard" for evaluating new and existing deconvolution methods. Below, we discuss these key challenges and how they can be addressed with the acquisition of new datasets and approaches to analysis.
Collapse
Affiliation(s)
- Sean K Maden
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Louise A Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | | | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Chen X, Yang M, Wang L, Wang Y, Tu J, Zhou X, Yuan X. Identification and in vitro and in vivo validation of the key role of GSDME in pyroptosis-related genes signature in hepatocellular carcinoma. BMC Cancer 2023; 23:411. [PMID: 37149620 PMCID: PMC10164321 DOI: 10.1186/s12885-023-10850-1] [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: 09/24/2022] [Accepted: 04/14/2023] [Indexed: 05/08/2023] Open
Abstract
We used pyroptosis-related genes to establish a risk-score model for prognostic prediction of liver hepatocellular carcinoma (LIHC) patients. A total of 52 pyroptosis-associated genes were identified. Then, data for 374 LIHC patients and 50 normal individuals were acquired from the TCGA database. Through gene expression analyses, differentially expressed genes (DEGs) were determined. The 13 pyroptosis-related genes (PRGs) confirmed as potential prognostic factors through univariate Cox regression analysis were entered into Lasso and multivariate Cox regression to build a PRGs prognostic signature, containing four PRGs (BAK1, GSDME, NLRP6, and NOD2) determined as independent prognostic factors. mRNA levels were evaluated by qRT-PCR, while overall survival (OS) rates were assessed by the Kaplan-Meier method. Enrichment analyses were done to establish the mechanisms associated with differential survival status of LIHC patients from a tumor immunology perspective. Additionally, a risk score determined by the prognostic model could divide LIHC patients into low- and high-risk groups using median risk score as cut-off. A prognostic nomogram, derived from the prognostic model and integrating clinical characteristics of patients, was constructed. The prognostic function of the model was also validated using GEO, ICGC cohorts, and online databases Kaplan-Meier Plotter. Small interfering RNA-mediated knockdown of GSDME, as well as lentivirus-mediated GSDME knockdown, were performed to validate that knockdown of GSDME markedly suppressed growth of HCC cells both in vivo and in vitro. Collectively, our study demonstrated a PRGs prognostic signature that had great clinical value in prognosis assessment.
Collapse
Affiliation(s)
- Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China
| | - Mu Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China
| | - Yuan Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China.
| | - Xiao Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei, China.
| |
Collapse
|
21
|
Zhang S, Zheng Y, Li X, Zhang S, Hu H, Kuang W. Cellular senescence-related gene signature as a valuable predictor of prognosis in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:3064-3093. [PMID: 37059592 DOI: 10.18632/aging.204658] [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: 02/06/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a lethal tumor. Its prognosis prediction remains a challenge. Meanwhile, cellular senescence, one of the hallmarks of cancer, and its related prognostic genes signature can provide critical information for clinical decision-making. METHOD Using bulk RNA sequencing and microarray data of HCC samples, we established a senescence score model via multi-machine learning algorithms to predict the prognosis of HCC. Single-cell and pseudo-time trajectory analyses were used to explore the hub genes of the senescence score model in HCC sample differentiation. RESULT A machine learning model based on cellular senescence gene expression profiles was identified in predicting HCC prognosis. The feasibility and accuracy of the senescence score model were confirmed in external validation and comparison with other models. Moreover, we analyzed the immune response, immune checkpoints, and sensitivity to immunotherapy drugs of HCC patients in different prognostic risk groups. Pseudo-time analyses identified four hub genes in HCC progression, including CDCA8, CENPA, SPC25, and TTK, and indicated related cellular senescence. CONCLUSIONS This study identified a prognostic model of HCC by cellular senescence-related gene expression and insight into novel potential targeted therapies.
Collapse
Affiliation(s)
- Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yilu Zheng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xinyu Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hao Hu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, The First Dongguan Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
22
|
Crosstalk between Mesenchymal Stem Cells and Cancer Stem Cells Reveals a Novel Stemness-Related Signature to Predict Prognosis and Immunotherapy Responses for Bladder Cancer Patients. Int J Mol Sci 2023; 24:ijms24054760. [PMID: 36902193 PMCID: PMC10003512 DOI: 10.3390/ijms24054760] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) and cancer stem cells (CSCs) maintain bladder cancer (BCa) stemness and facilitate the progression, metastasis, drug resistance, and prognosis. Therefore, we aimed to decipher the communication networks, develop a stemness-related signature (Stem. Sig.), and identify a potential therapeutic target. BCa single-cell RNA-seq datasets (GSE130001 and GSE146137) were used to identify MSCs and CSCs. Pseudotime analysis was performed by Monocle. Stem. Sig. was developed by analyzing the communication network and gene regulatory network (GRN) that were decoded by NicheNet and SCENIC, respectively. The molecular features of the Stem. Sig. were evaluated in TCGA-BLCA and two PD-(L)1 treated datasets (IMvigor210 and Rose2021UC). A prognostic model was constructed based on a 101 machine-learning framework. Functional assays were performed to evaluate the stem traits of the hub gene. Three subpopulations of MSCs and CSCs were first identified. Based on the communication network, the activated regulons were found by GRN and regarded as the Stem. Sig. Following unsupervised clustering, two molecular subclusters were identified and demonstrated distinct cancer stemness, prognosis, immunological TME, and response to immunotherapy. Two PD-(L)1 treated cohorts further validated the performance of Stem. Sig. in prognosis and immunotherapeutic response prediction. A prognostic model was then developed, and a high-risk score indicated a poor prognosis. Finally, the hub gene SLC2A3 was found exclusively upregulated in extracellular matrix-related CSCs, predicting prognosis, and shaping an immunosuppressive tumor microenvironment. Functional assays uncovered the stem traits of SLC2A3 in BCa by tumorsphere formation and western blotting. The Stem. Sig. derived from MSCs and CSCs can predict prognosis and response to immunotherapy for BCa. Besides, SLC2A3 may serve as a promising stemness target facilitating cancer effective management.
Collapse
|
23
|
Liu Y, Jian J, Zhang Y, Wang L, Liu X, Chen Z. Construction of cancer- associated fibroblasts related risk signature based on single-cell RNA-seq and bulk RNA-seq data in bladder urothelial carcinoma. Front Oncol 2023; 13:1170893. [PMID: 37124542 PMCID: PMC10140328 DOI: 10.3389/fonc.2023.1170893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
Background The ability of cancer-associated fibroblasts (CAFs) to encourage angiogenesis, tumor cell spread, and increase treatment resistance makes them pro-tumorigenic. We aimed to investigate the CAF signature in Bladder urothelial carcinoma (BLCA) and, for clinical application, to build a CAF-based risk signature to decipher the immune landscape and screen for suitable treatment BLCA samples. Methods CAF-related genes were discovered by superimposing CAF marker genes discovered from single-cell RNA-seq (scRNA-seq) data taken from the GEO database with CAF module genes discovered by weighted gene co-expression network analysis (WGCNA) using bulk RNA-seq data from TCGA. After identifying prognostic genes related with CAF using univariate Cox regression, Lasso regression was used to build a risk signature. With microarray data from the GEO database, prognostic characteristics were externally verified. For high and low CAF-risk categories, immune cells and immunotherapy responses were analyzed. Finally, a nomogram model based on the risk signature and prospective chemotherapeutic drugs were examined. Results Combining scRNA-seq and bulk-seq data analysis yielded a total of 124 CAF-related genes. LRP1, ANXA5, SERPINE2, ECM1, RBP1, GJA1, and FKBP10 were the seven BLCA prognostic genes that remained after univariate Cox regression and LASSO regression analyses. Then, based on these genes, prognostic characteristics were created and validated to predict survival in BLCA patients. Additionally, risk signature had a strong correlation with known CAF scores, stromal scores, and certain immune cells. The CAF-risk signature was identified as an independent prognostic factor for BLCA using multifactorial analysis, and its usefulness in predicting immunotherapy response was confirmed. Based on risk classification, we projected six highly sensitive anticancer medicines for the high-risk group. Conclusion The prognosis of BLCA may be accurately predicted using CAF-based risk signature. With a thorough understanding of the BLCA CAF-signature, it might be able to explain the BLCA patients' response to immunotherapy and identify a potential target for BLCA treatment.
Collapse
Affiliation(s)
- Yunxun Liu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jun Jian
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Ye Zhang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
- *Correspondence: Xiuheng Liu, ; Zhiyuan Chen,
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, China
- *Correspondence: Xiuheng Liu, ; Zhiyuan Chen,
| |
Collapse
|
24
|
Xia M, Wang S, Wang L, Mei Y, Tu Y, Gao L. The role of lactate metabolism-related LncRNAs in the prognosis, mutation, and tumor microenvironment of papillary thyroid cancer. Front Endocrinol (Lausanne) 2023; 14:1062317. [PMID: 37025405 PMCID: PMC10070953 DOI: 10.3389/fendo.2023.1062317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/07/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Lactate, a byproduct of glucose metabolism, is primarily utilized for gluconeogenesis and numerous cellular and organismal life processes. Interestingly, many studies have demonstrated a correlation between lactate metabolism and tumor development. However, the relationship between long non-coding RNAs (lncRNAs) and lactate metabolism in papillary thyroid cancer (PTC) remains to be explored. METHODS Lactate metabolism-related lncRNAs (LRLs) were obtained by differential expression and correlation analyses, and the risk model was further constructed by least absolute shrinkage and selection operator analysis (Lasso) and Cox analysis. Clinical, immune, tumor mutation, and enrichment analyses were performed based on the risk model. The expression level of six LRLs was tested using RT-PCR. RESULTS This study found several lncRNAs linked to lactate metabolism in both The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) datasets. Using Cox regression analysis, 303 lactate LRLs were found to be substantially associated with prognosis. Lasso was done on the TCGA cohort. Six LRLs were identified as independent predictive indicators for the development of a PTC prognostic risk model. The cohort was separated into two groups based on the median risk score (0.39717 -0.39771). Subsequently, Kaplan-Meier survival analysis and multivariate Cox regression analysis revealed that the high-risk group had a lower survival probability and that the risk score was an independent predictive factor of prognosis. In addition, a nomogram that can easily predict the 1-, 3-, and 5-year survival rates of PTC patients was established. Furthermore, the association between PTC prognostic factors and tumor microenvironment (TME), immune escape, as well as tumor somatic mutation status was investigated in high- and low-risk groups. Lastly, gene expression analysis was used to confirm the differential expression levels of the six LRLs. CONCLUSION In conclusion, we have constructed a prognostic model that can predict the prognosis, mutation status, and TME of PTC patients. The model may have great clinical significance in the comprehensive evaluation of PTC patients.
Collapse
Affiliation(s)
- Minqi Xia
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuo Wang
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Wang
- Department of Infection Prevention and Control Office, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Ling Gao,
| | - Yingna Mei
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Tu
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Ling Gao,
| |
Collapse
|
25
|
Fan Y, S Chan A, Zhu J, Yi Leung S, Fan X. A Bayesian model for identifying cancer subtypes from paired methylation profiles. Brief Bioinform 2022; 24:6961790. [PMID: 36575828 PMCID: PMC9851340 DOI: 10.1093/bib/bbac568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/19/2022] [Accepted: 11/22/2022] [Indexed: 12/29/2022] Open
Abstract
Aberrant DNA methylation is the most common molecular lesion that is crucial for the occurrence and development of cancer, but has thus far been underappreciated as a clinical tool for cancer classification, diagnosis or as a guide for therapeutic decisions. Partly, this has been due to a lack of proven algorithms that can use methylation data to stratify patients into clinically relevant risk groups and subtypes that are of prognostic importance. Here, we proposed a novel Bayesian model to capture the methylation signatures of different subtypes from paired normal and tumor methylation array data. Application of our model to synthetic and empirical data showed high clustering accuracy, and was able to identify the possible epigenetic cause of a cancer subtype.
Collapse
Affiliation(s)
- Yetian Fan
- School of Mathematics and Statistics, Liaoning University, Shenyang, 110036, China,Department of Statistics, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, China
| | - April S Chan
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jun Zhu
- Sema4, Stamford, CT, 06902, USA,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Suet Yi Leung
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xiaodan Fan
- Corresponding author: Xiaodan Fan, Department of Statistics, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR, China. E-mail:
| |
Collapse
|
26
|
Chen H, Yang W, Xue X, Li Y, Jin Z, Ji Z. Integrated Analysis Revealed an Inflammatory Cancer-Associated Fibroblast-Based Subtypes with Promising Implications in Predicting the Prognosis and Immunotherapeutic Response of Bladder Cancer Patients. Int J Mol Sci 2022; 23:ijms232415970. [PMID: 36555612 PMCID: PMC9781727 DOI: 10.3390/ijms232415970] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Inflammatory cancer-associated fibroblasts (iCAFs) are closely related to progression, anticancer therapeutic resistance, and poor prognosis of bladder cancer (BCa). However, the functional role of iCAFs in BCa has been poorly studied. In our study, two BCa scRNA-seq datasets (GSE130001 and GSE146137) were obtained and integrated by the Seurat pipeline. Based on reported markers (COL1A1 and PDGFRA), iCAFs were identified and the related signature of 278 markers was developed. Following unsupervised consensus clustering, two molecular subtypes of TCGA-BLCA were identified and characterized by distinct dysregulated cancer hallmarks, immunological tumor microenvironments, prognoses, responses to chemotherapy/immunotherapy, and stemness. Subsequently, the robustness of the signature-based clustering, in terms of prognosis and therapeutic response prediction, was validated in a GEO-meta cohort with seven independent GEO datasets of 519 BCa patients, and three immune checkpoint inhibitor (ICI)-treated cohorts. Considering the heterogeneity, re-clustering of iCAFs was performed and a subpopulation, named "LOXL2+ iCAFs", was identified. Co-culture CM derived from LOXL2 overexpression/silencing CAFs with T24 cells revealed that overexpression of LOXL2 in CAFs promoted while silencing LOXL2 inhibited the proliferation, migration, and invasion of T24 cells through IL32. Moreover, the positive correlation between LOXL2 and CD206, an M2 macrophage polarization marker, has been observed and validated. Collectively, integrated single-cell and bulk RNA sequencing analyses revealed an iCAF-related signature that can predict prognosis and response to immunotherapy for BCa. Additionally, the hub gene LOXL2 may serve as a promising target for BCa treatment.
Collapse
|
27
|
Yu H, Sfakianos JP, Wang L, Hu Y, Daza J, Galsky MD, Sandhu HS, Elemento O, Faltas BM, Farkas AM, Bhardwaj N, Zhu J, Mulholland DJ. Tumor-Infiltrating Myeloid Cells Confer De Novo Resistance to PD-L1 Blockade through EMT-Stromal and Tgfβ-Dependent Mechanisms. Mol Cancer Ther 2022; 21:1729-1741. [PMID: 36129800 PMCID: PMC9706595 DOI: 10.1158/1535-7163.mct-22-0130] [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: 02/23/2022] [Revised: 05/31/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022]
Abstract
SIGNIFICANCE Most patients with bladder cancer do not respond to ICB targeting of the PD-L1 signaling axis. Our modeling applied a de novo resistance signature to show that tumor-infiltrating myeloid cells promote poor treatment response in a TGFβ-dependent mechanism.
Collapse
Affiliation(s)
| | - John P. Sfakianos
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Li Wang
- Sema4, Stamford, CT 06902 USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yang Hu
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10065
| | - Jorge Daza
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Matthew D. Galsky
- Division of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, 10029
| | - Harkirat S. Sandhu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, 10029
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10065
| | - Bishoy M. Faltas
- Departments of Medicine, Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, 10065
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10065
| | | | | | - Jun Zhu
- Sema4, Stamford, CT 06902 USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - David J. Mulholland
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, 10029
| |
Collapse
|
28
|
Cai J, Ji Z, Wu J, Chen L, Zheng D, Chen Y, Zhang X, Xie W, Huang J, Chen M, Lin R, Lin W, Chen Y, Li Z. Development and validation of a novel endoplasmic reticulum stress-related lncRNA prognostic signature and candidate drugs in breast cancer. Front Genet 2022; 13:949314. [PMID: 36092873 PMCID: PMC9452962 DOI: 10.3389/fgene.2022.949314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/27/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer (BC), the most common malignancy in women, has a high cancer-related mortality. Endoplasmic reticulum stress (ERS), a response to the accumulation of unfolded proteins, has emerging roles in tumorigenesis, including invasion, metastasis, immune escape, etc. However, few studies have focused on the correlation between ERS with long non-coding RNAs (lncRNAs) in BC. We attempted to construct an ERS-related lncRNA prognostic signature and study its value in BC from tumor mutational burden (TMB), tumor immune microenvironment (TIME), cluster, clinical treatment, and so on. In the present study, transcriptomic and clinical data of BC patients were extracted from The Cancer Genome Atlas (TCGA) database. Correlation test, Cox regression analysis, least absolute shrinkage, and selection operator (LASSO) method were performed to determine an ERS-related lncRNA prognostic signature. Survival and predictive performance were analyzed according to Kaplan-Meier curves and receiver operating characteristic (ROC) curves, while nomograms and calibration curves were established. Then, an enrichment analysis was performed to study the functions and biological processes of ERS-related lncRNAs. TMB and TIME were also analyzed to assess the mutational status and immune status. Additionally, by using consensus cluster analysis, we compared differences among tumor subtypes. Drug sensitivity analysis and immunologic efficacy evaluations were performed together for further exploration. We identified a novel prognostic signature consisting of 9 ERS-related lncRNAs. High-risk patients had worse prognoses. The signature had a good predictive performance as an independent prognostic indicator and was significantly associated with clinicopathological characteristics. Enrichment analysis showed that metabolic pathways were enriched in high-risk patients, while immune pathways were more active in low-risk patients. Low-risk patients had lower TMB, higher immune scores, and stronger immune functions. Cluster analysis clarified that cluster 2 had the most active immune functions and was sensitive to more drugs, which may have the best clinical immunological efficacy. A clinical efficacy evaluation revealed that patients in the low-risk group may benefit more from chemotherapy, targeted therapy, and immunotherapy. The novel signature has significant clinical implications in prognosis prediction for BC. Our study clarifies that there is a potential connection between the ERS-related lncRNAs and BC, which may provide new treatment guidelines for BC.
Collapse
Affiliation(s)
- Jiehui Cai
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zeqi Ji
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jinyao Wu
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | | | | | | | | | | | | | | | | | | | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zhiyang Li
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
29
|
Yu H, Tuminello S, Alpert N, van Gerwen M, Yoo S, Mulholland DJ, Aaronson SA, Donovan M, Oh WK, Gong Y, Wang L, Zhu J, Taioli E. Global DNA methylation of WTC prostate cancer tissues show signature differences compared to non-exposed cases. Carcinogenesis 2022; 43:528-537. [PMID: 35239955 PMCID: PMC9234756 DOI: 10.1093/carcin/bgac025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/08/2022] [Accepted: 03/02/2022] [Indexed: 12/31/2022] Open
Abstract
There is increased incidence of prostate cancer (PC) among World Trade Center (WTC)-exposed responders and community members, with preliminary evidence suggestive of more aggressive disease. While previous research is supportive of differences in DNA methylation and gene expression as a consequence of WTC exposure, as measured in blood of healthy individuals, the epigenetics of WTC PC tissues has yet to be explored. Patients were recruited from the World Trade Center Health Program. Non-WTC PC samples were frequency matched on age, race/ethnicity and Gleason score. Bisulfite-treated DNA was extracted from tumor tissue blocks and used to assess global DNA methylation with the MethylationEPIC BeadChip. Differential and pathway enrichment analyses were conducted. RNA from the same tumor blocks was used for gene expression analysis to further support DNA methylation findings. Methylation data were generated for 28 samples (13 WTC and 15 non-WTC). Statistically significant differences in methylation were observed for 3,586 genes; on average WTC samples were statistically significantly more hypermethylated (P = 0.04131). Pathway enrichment analysis revealed hypermethylation in epithelial mesenchymal transition (EMT), hypoxia, mitotic spindle, TNFA signaling via NFKB, WNT signaling, and TGF beta signaling pathways in WTC compared to non-WTC samples. The androgen response, G2M and MYC target pathways were hypomethylated. These results correlated well with RNA gene expression. In conclusion, long-term epigenic changes associated with WTC dust exposure were observed in PC tissues. These occurred in genes of critical pathways, likely increasing prostate tumorigenesis potential. This warrants analysis of larger WTC groups and other cancer types.
Collapse
Affiliation(s)
- Haocheng Yu
- Sema4, a Mount Sinai venture, Stamford, CT, USA
| | - Stephanie Tuminello
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Population Health, New York University Langone Health, New York, NY, USA
| | - Naomi Alpert
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maaike van Gerwen
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - David J Mulholland
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Donovan
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William K Oh
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yixuan Gong
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Wang
- Sema4, a Mount Sinai venture, Stamford, CT, USA
- Icahn Institute for Data Science and Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jun Zhu
- Sema4, a Mount Sinai venture, Stamford, CT, USA
- Icahn Institute for Data Science and Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emanuela Taioli
- Institute for Translational Epidemiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NYUSA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Yao J, Liu Y, Yang J, Li M, Li S, Zhang B, Yang R, Zhang Y, Cui X, Feng C. Single-Cell Sequencing Reveals that DBI is the Key Gene and Potential Therapeutic Target in Quiescent Bladder Cancer Stem Cells. Front Genet 2022; 13:904536. [PMID: 35769986 PMCID: PMC9235029 DOI: 10.3389/fgene.2022.904536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Drug resistance and recurrence often develop during the treatment of muscle-invasive bladder cancer (MIBC). The existence of cancer stem cells (CSCs) in MIBC makes the formulation of effective treatment strategies extremely challenging. We aimed to use single-cell RNA sequencing approaches to identify CSCs and evaluate their molecular characteristics and to discover possible therapeutic measures. Methods: GEO data sets GSE130001 and GSE146137 were used to construct an expression matrix. After cells were identified by type, malignant epithelial cells inferred by InferCNV were extracted for stemness evaluation. The subset of cells with the highest stemness was subjected to weighted gene coexpression network analysis (WGCNA) and pseudotime analysis to identify key genes. In addition, we predicted drug sensitivity relationships for key genes in CTD and predicted the correlation between drugs and survival through siGDC. Results: We found that there were some CSCs in MIBC samples. The CSC population was heterogeneous during tumor development and was divided into quiescent and proliferating CSCs. We identified DBI as the key gene in quiescent CSCs. Analysis of a TCGA data set showed that higher DBI expression indicated higher histological grade. In addition, we predicted that acetaminophen can reduce DBI expression, thereby reducing the stemness of CSCs. Thus, we identified a potential new use of acetaminophen. Conclusion: We systematically explored CSCs in tumors and determined that DBI may be a key gene and potential therapeutic target in quiescent CSCs. In addition, we confirmed that acetaminophen may be a candidate drug targeting CSCs, improving our understanding of CSC-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Jiaxi Yao
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yue Liu
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jitao Yang
- Department of Urology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mengling Li
- Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Simin Li
- Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Rui Yang
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuchong Zhang
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaoyu Cui
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Shenyang, China
- *Correspondence: Xiaoyu Cui, ; ChunQing Feng,
| | - ChunQing Feng
- Department of Urology Surgery, The Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
- *Correspondence: Xiaoyu Cui, ; ChunQing Feng,
| |
Collapse
|
31
|
Inflammatory Response-Related Long Non-Coding RNA Signature Predicts the Prognosis of Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:9917244. [PMID: 35342418 PMCID: PMC8947866 DOI: 10.1155/2022/9917244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/22/2022] [Indexed: 01/08/2023]
Abstract
Background Hepatocellular carcinoma (HCC) is a high mortality malignant tumor with genetic and phenotypic heterogeneity, making predicting prognosis challenging. Meanwhile, the inflammatory response is an indispensable player in the tumorigenesis process and regulates the tumor microenvironment, which can affect the prognosis of tumor patients. Methods Using HCC samples in the TCGA-LIHC dataset, we explored lncRNA expression profiles associated with the inflammatory response. The inflammatory response-related lncRNA signature was constructed by univariate Cox regression, LASSO regression, and multivariate Cox regression methods based on inflammatory response-related differentially expressed lncRNAs in HCC. Results Seven inflammatory response-related lncRNA signatures were identified in predicting HCC prognosis. Kaplan–Meier (K-M) survival analysis indicated that high-risk group HCC patients were associated with poor prognosis. The utility of the inflammatory response-related lncRNA signatures was proved by the AUC and DCA analysis. The nomogram further confirmed the accuracy of the novel signature in predicting HCC patients' prognoses. In validation, our novel signature is more accurate than traditional clinicopathological performance for prognosis prediction of HCC patients. GSEA analysis further elucidated the underlying mechanisms and pathways of HCC progression in the low- and high-risk groups. Moreover, immune cells infiltration responses and immune function analyses revealed a significant difference between high- and low-risk groups in cytolytic activity, MHC class I, type I INF response, type II INF response, inflammation-promoting, and T cell coinhibition. Finally, HHLA2, NRP1, CD276, TNFRSF9, TNFSF4, CD80, and VTCN1 were expressed higher in high-risk groups in the immune checkpoint analysis. Conclusions A novel inflammatory response-related lncRNA signature (AC145207.5, POLHAS1, AL928654.1, MKLN1AS, AL031985.3, PRRT3AS1, and AC023157.2) is capable of predicting the prognosis of HCC patients and providing new immune targeted therapies insight.
Collapse
|
32
|
Zhang S, Li X, Zhang X, Zhang S, Tang C, Kuang W. The Pyroptosis-Related Gene Signature Predicts the Prognosis of Hepatocellular Carcinoma. Front Mol Biosci 2022; 8:781427. [PMID: 35047554 PMCID: PMC8762168 DOI: 10.3389/fmolb.2021.781427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
Objective: Hepatocellular carcinoma (HCC) is a genetically and phenotypically heterogeneous tumor, and the prediction of its prognosis remains a challenge. In the past decade, studies elucidating the mechanisms that induce tumor cell pyroptosis has rapidly increased. The elucidation of their mechanisms is essential for the clinical development optimal application of anti-hepatocellular carcinoma therapeutics. Methods: Based on the different expression profiles of pyroptosis-related genes in HCC, we constructed a LASSO Cox regression pyroptosis-related genes signature that could more accurately predict the prognosis of HCC patients. Results: We identified seven pyroptosis-related genes signature (BAK1, CHMP4B, GSDMC, NLRP6, NOD2, PLCG1, SCAF11) in predicting the prognosis of HCC patients. Kaplan Meier survival analysis showed that the pyroptosis-related high-risk gene signature was associated with poor prognosis HCC patients. Moreover, the pyroptosis-related genes signature performed well in the survival analysis and ICGC validation group. The hybrid nomogram and calibration curve further demonstrated their feasibility and accuracy for predicting the prognosis of HCC patients. Meanwhile, the evaluation revealed that our novel signature predicted the prognosis of HCC patients more accurately than traditional clinicopathological features. GSEA analysis further revealed the novel signature associated mechanisms of immunity response in high-risk groups. Moreover, analysis of immune cell subsets with relevant functions revealed significant differences in aDCs, APC co-stimulation, CCR, check-point, iDCs, Macrophages, MHC class-I, Treg, and type II INF response between high- and low-risk groups. Finally, the expression of Immune checkpoints was enhanced in high-risk group, and m6A-related modifications were expressed differently between low- and high-risk groups. Conclusion: The novel pyroptosis-related genes signature can predict the prognosis of patients with HCC and insight into new cell death targeted therapies.
Collapse
Affiliation(s)
- Shuqiao Zhang
- First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyu Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shijun Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chunzhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
33
|
Perez MF, Sarkies P. Malignancy and NF-κB signalling strengthen coordination between expression of mitochondrial and nuclear-encoded oxidative phosphorylation genes. Genome Biol 2021; 22:328. [PMID: 34857014 PMCID: PMC8638269 DOI: 10.1186/s13059-021-02541-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Mitochondria are ancient endosymbiotic organelles crucial to eukaryotic growth and metabolism. The mammalian mitochondrial genome encodes for 13 mitochondrial proteins, and the remaining mitochondrial proteins are encoded by the nuclear genome. Little is known about how coordination between the expression of the two sets of genes is achieved. RESULTS Correlation analysis of RNA-seq expression data from large publicly available datasets is a common method to leverage genetic diversity to infer gene co-expression modules. Here we use this method to investigate nuclear-mitochondrial gene expression coordination. We identify a pitfall in correlation analysis that results from the large variation in the proportion of transcripts from the mitochondrial genome in RNA-seq data. Commonly used normalisation techniques based on total read counts, such as FPKM or TPM, produce artefactual negative correlations between mitochondrial- and nuclear-encoded transcripts. This also results in artefactual correlations between pairs of nuclear-encoded genes, with important consequences for inferring co-expression modules beyond mitochondria. We show that these effects can be overcome by normalizing using the median-ratio normalisation (MRN) or trimmed mean of M values (TMM) methods. Using these normalisations, we find only weak and inconsistent correlations between mitochondrial and nuclear-encoded mitochondrial genes in the majority of healthy human tissues from the GTEx database. CONCLUSIONS We show that a subset of healthy tissues with high expression of NF-κB show significant coordination, suggesting a role for NF-κB in ensuring balanced expression between mitochondrial and nuclear genes. Contrastingly, most cancer types show robust coordination of nuclear and mitochondrial OXPHOS gene expression, identifying this as a feature of gene regulation in cancer.
Collapse
Affiliation(s)
- Marcos Francisco Perez
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| | - Peter Sarkies
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
34
|
Zhang S, Li X, Tang C, Kuang W. Inflammation-Related Long Non-Coding RNA Signature Predicts the Prognosis of Gastric Carcinoma. Front Genet 2021; 12:736766. [PMID: 34819945 PMCID: PMC8607501 DOI: 10.3389/fgene.2021.736766] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Gastric carcinoma (GC) is a molecularly and phenotypically highly heterogeneous disease, making the prognostic prediction challenging. On the other hand, Inflammation as part of the active cross-talk between the tumor and the host in the tumor or its microenvironment could affect prognosis. Method: We established a prognostic multi lncRNAs signature that could better predict the prognosis of GC patients based on inflammation-related differentially expressed lncRNAs in GC. Results: We identified 10 differently expressed lncRNAs related to inflammation associated with GC prognosis. Kaplan-Meier survival analysis demonstrated that high-risk inflammation-related lncRNAs signature was related to poor prognosis of GC. Moreover, the inflammation-related lncRNAs signature had an AUC of 0.788, proving their utility in predicting GC prognosis. Indeed, our risk signature is more precise in predicting the prognosis of GC patients than traditional clinicopathological manifestations. Immune and tumor-related pathways for individuals in the low and high-risk groups were further revealed by GSEA. Moreover, TCGA based analysis revealed significant differences in HLA, MHC class-I, cytolytic activity, parainflammation, co-stimulation of APC, type II INF response, and type I INF response between the two risk groups. Immune checkpoints revealed CD86, TNFSF18, CD200, and LAIR1 were differently expressed between lowand high-risk groups. Conclusion: A novel inflammation-related lncRNAs (AC015660.1, LINC01094, AL512506.1, AC124067.2, AC016737.1, AL136115.1, AP000695.1, AC104695.3, LINC00449, AC090772.1) signature may provide insight into the new therapies and prognosis prediction for GC patients.
Collapse
Affiliation(s)
- ShuQiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - XinYu Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - ChunZhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - WeiHong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
35
|
Integrative Transcriptome Profiling Reveals SKA3 as a Novel Prognostic Marker in Non-Muscle Invasive Bladder Cancer. Cancers (Basel) 2021; 13:cancers13184673. [PMID: 34572901 PMCID: PMC8470398 DOI: 10.3390/cancers13184673] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 01/03/2023] Open
Abstract
Approximately 80% of all new bladder cancer patients are diagnosed with non-muscle invasive bladder cancer (NMIBC). However, approximately 15% of them progress to muscle-invasive bladder cancer (MIBC), for which prognosis is poor. The current study aimed to improve diagnostic accuracy associated with clinical outcomes in NMIBC patients. Nevertheless, it has been challenging to identify molecular biomarkers that accurately predict MIBC progression because this disease is complex and heterogeneous. Through integrative transcriptome profiling, we showed that high SKA3 expression is associated with poor clinical outcomes and MIBC progression. We performed RNA sequencing on human tumor tissues to identify candidate biomarkers in NMIBC. We then selected genes with prognostic significance by analyzing public datasets from multiple cohorts of bladder cancer patients. We found that SKA3 was associated with NMIBC pathophysiology and poor survival. We analyzed public single-cell RNA-sequencing (scRNA-seq) data for bladder cancer to dissect transcriptional tumor heterogeneity. SKA3 was expressed in an epithelial cell subpopulation expressing genes regulating the cell cycle. Knockdown experiments confirmed that SKA3 promotes bladder cancer cell proliferation by accelerating G2/M transition. Hence, SKA3 is a new prognostic marker for predicting NMIBC progression. Its inhibition could form part of a novel treatment lowering the probability of bladder cancer progression.
Collapse
|
36
|
Wang W, Wang L, She J, Zhu J. Examining heterogeneity of stromal cells in tumor microenvironment based on pan-cancer single-cell RNA sequencing data. Cancer Biol Med 2021; 19:j.issn.2095-3941.2020.0762. [PMID: 34398535 PMCID: PMC8763007 DOI: 10.20892/j.issn.2095-3941.2020.0762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/05/2021] [Indexed: 11/28/2022] Open
Abstract
Tumor tissues contain both tumor and non-tumor cells, which include infiltrated immune cells and stromal cells, collectively called the tumor microenvironment (TME). Single-cell RNA sequencing (scRNAseq) enables the examination of heterogeneity of tumor cells and TME. In this review, we examined scRNAseq datasets for multiple cancer types and evaluated the heterogeneity of major cell type composition in different cancer types. We further showed that endothelial cells and fibroblasts/myofibroblasts in different cancer types can be classified into common subtypes, and the subtype composition is clearly associated with cancer characteristic and therapy response.
Collapse
Affiliation(s)
- Wenhui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Li Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Sema4, a Mount Sinai venture, Stamford, CT 06902, USA
| | - Junjun She
- First Affiliate Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Sema4, a Mount Sinai venture, Stamford, CT 06902, USA
- First Affiliate Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
37
|
Ma Q, Chen Y, Xiao F, Hao Y, Song Z, Zhang J, Okuda K, Um SW, Silva M, Shimada Y, Si C, Liang C. A signature of estimate-stromal-immune score-based genes associated with the prognosis of lung adenocarcinoma. Transl Lung Cancer Res 2021; 10:1484-1500. [PMID: 33889524 PMCID: PMC8044489 DOI: 10.21037/tlcr-21-223] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Immune and stromal component evaluation is necessary to establish accurate prognostic markers for the prediction of clinical outcomes in lung adenocarcinoma (LUAD). We aimed to develop a gene signature based on the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE)-stromal-immune score in LUAD. Methods The transcriptomic profiles of patients with LUAD were obtained from The Cancer Genome Atlas (TCGA), and the immune and stromal scores were derived using the ESTIMATE algorithm. The prognostic signature genes were selected from the differentially expressed genes (DEGs) using the robust partial likelihood-based cox proportional hazards regression method. The negative log-likelihood and the Akaike Information Criterion (AIC) were used to identify the optimal gene signature. The validation was carried out in 2 independent datasets from the Gene Expression Omnibus (GSE68571 and GSE72094). Results Patients with high ESTIMATE, stromal, and immune scores had better overall survivals (P=0.0035, P=0.066, and P=0.0077). The expression of thirty-seven genes was related to ESTIMATE-stromal-immune score. A risk stratification model was developed based on a gene signature containing CD74, JCHAIN, and PTGDS. The ESTIMATE-stromal-immune risk score was revealed to be a prognostic factor (P=0.009) after multivariate analysis. Four groups were classified based on this risk stratification model, yielding increasing survival outcomes (log-rank test, P=0.0051). This risk stratification model and other clinicopathological factors were combined to generate a nomogram. The calibration curves showed perfect agreement between the nomogram-predicted outcomes and those actually observed. Similar observations were made in 2 independent cohorts. Conclusions The gene signature based on the ESTIMATE-stromal-immune score could predict the prognosis of patients with LUAD.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yang Chen
- Department of Biochemistry and Molecular Biology, The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yang Hao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhiyi Song
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Katsuhiro Okuda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sang-Won Um
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mario Silva
- Section of Scienze Radiologiche, Department of Medicine and Surgery (DiMeC), University of Parma, Parma, Italy
| | - Yoshihisa Shimada
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Chaozeng Si
- Department of Information Management, China-Japan Friendship Hospital, Beijing, China
| | - Chaoyang Liang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
38
|
Hu Z, Cunnea P, Zhong Z, Lu H, Osagie OI, Campo L, Artibani M, Nixon K, Ploski J, Santana Gonzalez L, Alsaadi A, Wietek N, Damato S, Dhar S, Blagden SP, Yau C, Hester J, Albukhari A, Aboagye EO, Fotopoulou C, Ahmed A. The Oxford Classic Links Epithelial-to-Mesenchymal Transition to Immunosuppression in Poor Prognosis Ovarian Cancers. Clin Cancer Res 2021; 27:1570-1579. [PMID: 33446563 DOI: 10.1158/1078-0432.ccr-20-2782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/03/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Using RNA sequencing, we recently developed the 52-gene-based Oxford classifier of carcinoma of the ovary (Oxford Classic, OxC) for molecular stratification of serous ovarian cancers (SOCs) based on the molecular profiles of their cell of origin in the fallopian tube epithelium. Here, we developed a 52-gene NanoString panel for the OxC to test the robustness of the classifier. EXPERIMENTAL DESIGN We measured the expression of the 52 genes in an independent cohort of prospectively collected SOC samples (n = 150) from a homogenous cohort who were treated with maximal debulking surgery and chemotherapy. We performed data mining of published expression profiles of SOCs and validated the classifier results on tissue arrays comprising 137 SOCs. RESULTS We found evidence of profound nongenetic heterogeneity in SOCs. Approximately 20% of SOCs were classified as epithelial-to-mesenchymal transition-high (EMT-high) tumors, which were associated with poor survival. This was independent of established prognostic factors, such as tumor stage, tumor grade, and residual disease after surgery (HR, 3.3; P = 0.02). Mining expression data of 593 patients revealed a significant association between the EMT scores of tumors and the estimated fraction of alternatively activated macrophages (M2; P < 0.0001), suggesting a mechanistic link between immunosuppression and poor prognosis in EMT-high tumors. CONCLUSIONS The OxC-defined EMT-high SOCs carry particularly poor prognosis independent of established clinical parameters. These tumors are associated with high frequency of immunosuppressive macrophages, suggesting a potential therapeutic target to improve clinical outcome.
Collapse
Affiliation(s)
- Zhiyuan Hu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom
| | - Paula Cunnea
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, England, United Kingdom
| | - Zhe Zhong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom.,School of Life Science, Peking University, Beijing, P.R. China
| | - Haonan Lu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, England, United Kingdom.,Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, England, United Kingdom
| | - Oloruntoba I Osagie
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom
| | - Leticia Campo
- Department of Oncology, University of Oxford, Oxford, England, United Kingdom
| | - Mara Artibani
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom.,Gene Regulatory Networks in Development and Disease Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, England, United Kingdom
| | - Katherine Nixon
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, England, United Kingdom
| | - Jennifer Ploski
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, England, United Kingdom
| | - Laura Santana Gonzalez
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom
| | - Abdulkhaliq Alsaadi
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom
| | - Nina Wietek
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom.,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom
| | - Stephen Damato
- Department of Histopathology, Oxford University Hospitals, Oxford, England, United Kingdom
| | - Sunanda Dhar
- Department of Histopathology, Oxford University Hospitals, Oxford, England, United Kingdom
| | - Sarah P Blagden
- Department of Oncology, University of Oxford, Oxford, England, United Kingdom
| | - Christopher Yau
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, England, United Kingdom.,Alan Turing Institute, London, England, United Kingdom
| | - Joanna Hester
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, England, United Kingdom
| | - Ashwag Albukhari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Eric O Aboagye
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, England, United Kingdom
| | - Christina Fotopoulou
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, England, United Kingdom.
| | - Ahmed Ahmed
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, United Kingdom. .,Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, England, United Kingdom
| |
Collapse
|
39
|
Chen Z, Wu A. Progress and challenge for computational quantification of tissue immune cells. Brief Bioinform 2021; 22:6065002. [PMID: 33401306 DOI: 10.1093/bib/bbaa358] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 11/07/2020] [Indexed: 12/28/2022] Open
Abstract
Tissue immune cells have long been recognized as important regulators for the maintenance of balance in the body system. Quantification of the abundance of different immune cells will provide enhanced understanding of the correlation between immune cells and normal or abnormal situations. Currently, computational methods to predict tissue immune cell compositions from bulk transcriptomes have been largely developed. Therefore, summarizing the advantages and disadvantages is appropriate. In addition, an examination of the challenges and possible solutions for these computational models will assist the development of this field. The common hypothesis of these models is that the expression of signature genes for immune cell types might represent the proportion of immune cells that contribute to the tissue transcriptome. In general, we grouped all reported tools into three groups, including reference-free, reference-based scoring and reference-based deconvolution methods. In this review, a summary of all the currently reported computational immune cell quantification tools and their applications, limitations, and perspectives are presented. Furthermore, some critical problems are found that have limited the performance and application of these models, including inadequate immune cell type, the collinearity problem, the impact of the tissue environment on the immune cell expression level, and the deficiency of standard datasets for model validation. To address these issues, tissue specific training datasets that include all known immune cells, a hierarchical computational framework, and benchmark datasets including both tissue expression profiles and the abundances of all the immune cells are proposed to further promote the development of this field.
Collapse
Affiliation(s)
- Ziyi Chen
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Jiangsu, Suzhou, China
| | - Aiping Wu
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Jiangsu, Suzhou, China
| |
Collapse
|
40
|
Haider S, Tyekucheva S, Prandi D, Fox NS, Ahn J, Xu AW, Pantazi A, Park PJ, Laird PW, Sander C, Wang W, Demichelis F, Loda M, Boutros PC. Systematic Assessment of Tumor Purity and Its Clinical Implications. JCO Precis Oncol 2020; 4:PO.20.00016. [PMID: 33015524 PMCID: PMC7529507 DOI: 10.1200/po.20.00016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2020] [Indexed: 02/03/2023] Open
Abstract
PURPOSE The tumor microenvironment is complex, comprising heterogeneous cellular populations. As molecular profiles are frequently generated using bulk tissue sections, they represent an admixture of multiple cell types (including immune, stromal, and cancer cells) interacting with each other. Therefore, these molecular profiles are confounded by signals emanating from many cell types. Accurate assessment of residual cancer cell fraction is crucial for parameterization and interpretation of genomic analyses, as well as for accurately interpreting the clinical properties of the tumor. MATERIALS AND METHODS To benchmark cancer cell fraction estimation methods, 10 estimators were applied to a clinical cohort of 333 patients with prostate cancer. These methods include gold-standard multiobserver pathology estimates, as well as estimates inferred from genome, epigenome, and transcriptome data. In addition, two methods based on genomic and transcriptomic profiles were used to quantify tumor purity in 4,497 tumors across 12 cancer types. Bulk mRNA and microRNA profiles were subject to in silico deconvolution to estimate cancer cell-specific mRNA and microRNA profiles. RESULTS We present a systematic comparison of 10 tumor purity estimation methods on a cohort of 333 prostate tumors. We quantify variation among purity estimation methods and demonstrate how this influences interpretation of clinico-genomic analyses. Our data show poor concordance between pathologic and molecular purity estimates, necessitating caution when interpreting molecular results. Limited concordance between DNA- and mRNA-derived purity estimates remained a general pan-cancer phenomenon when tested in an additional 4,497 tumors spanning 12 cancer types. CONCLUSION The choice of tumor purity estimation method may have a profound impact on the interpretation of genomic assays. Taken together, these data highlight the need for improved assessment of tumor purity and quantitation of its influences on the molecular hallmarks of cancers.
Collapse
Affiliation(s)
- Syed Haider
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Svitlana Tyekucheva
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Davide Prandi
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Natalie S. Fox
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, Washington, DC
| | - Andrew Wei Xu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | | | - Peter J. Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | | | - Chris Sander
- cBio Center, Dana-Farber Cancer Institute, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Wenyi Wang
- The University of Texas MD Anderson Cancer Center Department of Bioinformatics and Computational Biology, Houston
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
- Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY
| | - Massimo Loda
- Department of Pathology, Weill Medical College of Cornell University, New York, NY
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, CA
- Department of Urology, University of California, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA
- Institute for Precision Health, University of California, Los Angeles, CA
| | - The Cancer Genome Atlas Research Network
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, Washington, DC
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
- Brigham and Women’s Hospital, Boston, MA
- Van Andel Research Institute, Grand Rapids, MI
- cBio Center, Dana-Farber Cancer Institute, Boston, MA
- Department of Cell Biology, Harvard Medical School, Boston, MA
- The University of Texas MD Anderson Cancer Center Department of Bioinformatics and Computational Biology, Houston
- Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY
- Department of Pathology, Weill Medical College of Cornell University, New York, NY
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA
- Department of Human Genetics, University of California, Los Angeles, CA
- Department of Urology, University of California, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA
- Institute for Precision Health, University of California, Los Angeles, CA
| |
Collapse
|