1
|
Zheng Y, Peng Y, Gao Y, Yang G, Jiang Y, Zhang G, Wang L, Yu J, Huang Y, Wei Z, Liu J. Identification and dissection of prostate cancer grounded on fatty acid metabolism-correlative features for predicting prognosis and assisting immunotherapy. Comput Biol Chem 2024; 115:108323. [PMID: 39742702 DOI: 10.1016/j.compbiolchem.2024.108323] [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: 08/28/2024] [Revised: 11/24/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Fatty acid metabolism (FAM) plays a critical role in tumor progression and therapeutic resistance by enhancing lipid biosynthesis, storage, and catabolism. Dysregulated FAM is a hallmark of prostate cancer (PCa), enabling cancer cells to adapt to extracellular signals and metabolic changes, with the tumor microenvironment (TME) playing a key role. However, the prognostic significance of FAM in PCa remains unexplored. METHODS We analyzed 309 FAM-related genes to develop a prognostic model using least absolute shrinkage and selection operator (LASSO) regression based on The Cancer Genome Atlas (TCGA) database. This model stratified PCa patients into high- and low-risk groups and was validated using the Gene Expression Omnibus (GEO) database. We constructed a nomogram incorporating risk score, clinical variables (T and N stage, Gleason score, age), and assessed its performance with calibration curves. The associations between risk score, tumor mutation burden (TMB), immune checkpoint inhibitors (ICIs), and TME features were also examined. Finally, a hub gene was identified via protein-protein interaction (PPI) networks and validated. RESULTS The risk score was an independent prognostic factor for PCa. High-risk patients showed worse survival outcomes but were more responsive to immunotherapy, chemotherapy, and targeted therapies. A core gene with high expression correlated with poor prognosis, unfavorable clinicopathological features, and immune cell infiltration. CONCLUSION These findings reveal the prognostic importance of FAM in PCa, providing novel insights into prognosis and potential therapeutic targets for PCa management.
Collapse
Affiliation(s)
- Yongbo Zheng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yueqiang Peng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yingying Gao
- Department of Clinical Laboratory, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 401320, China
| | - Guo Yang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yu Jiang
- Department of Urology, The First Affiliated Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Gaojie Zhang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Linfeng Wang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Jiang Yu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yong Huang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Ziling Wei
- College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jiayu Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China.
| |
Collapse
|
2
|
Angel MR, Séguin B, Löhr CV, Beer TM, Feliciano J, Ramsey SA, Thomas GV. Comparative Transcriptomes of Canine and Human Prostate Cancers Identify Mediators of Castration Resistance. Vet Comp Oncol 2024; 22:629-640. [PMID: 39375962 DOI: 10.1111/vco.13017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 10/09/2024]
Abstract
Prostate cancer continues to be one of the most lethal cancers in men. While androgen deprivation therapy is initially effective in treating prostate cancer, most cases of advanced prostate cancer eventually progress to castration-resistant prostate cancer (CRPC), which is incurable. Similarly, the most aggressive form of prostatic carcinoma occurs in dogs that have been castrated. To identify molecular similarities between canine prostate cancer and human CRPC, we performed a comparative analysis of gene expression profiles. Through this transcriptomic analysis, we found that prostatic carcinoma in castrated dogs demonstrates an androgen-indifferent phenotype, characterised by low-androgen receptor and neuroendocrine-associated genes. Notably, we identified two genes, ISG15 and AZGP1, that were consistently up- and down-regulated, respectively, in both canine prostatic carcinoma and human CRPC. Additionally, we identified several other genes, including GPX3, S100P and IFITM1, that exhibited similar expression patterns in both species. Protein-protein interaction network analysis demonstrated that these five genes were part of a larger network of interferon-induced genes, suggesting that they may act together in signalling pathways that are disrupted in prostate cancer. Accordingly, our findings suggest that the interferon pathway may play a role in the development and progression of CRPC in both dogs and humans and chart a new therapeutic approach.
Collapse
Affiliation(s)
- Marcela Riveros Angel
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Bernard Séguin
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Christiane V Löhr
- College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - John Feliciano
- Veterinary Diagnostics & Imaging Consultants, Tualatin, Oregon, USA
| | - Stephen A Ramsey
- College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, USA
- College of Engineering, Oregon State University, Corvallis, Oregon, USA
| | - George V Thomas
- Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
3
|
Lin X, Liu J, Zhang N, Zhou D, Liu Y. Decoding the immune microenvironment: unveiling CD8 + T cell-related biomarkers and developing a prognostic signature for personalized glioma treatment. Cancer Cell Int 2024; 24:331. [PMID: 39354483 PMCID: PMC11443942 DOI: 10.1186/s12935-024-03517-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/25/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Gliomas are aggressive brain tumors with poor prognosis. Understanding the tumor immune microenvironment (TIME) in gliomas is essential for developing effective immunotherapies. This study aimed to identify TIME-related biomarkers in glioma using bioinformatic analysis of RNA-seq data. METHODS In this study, we employed weighted gene co-expression network analysis (WGCNA) on bulk RNA-seq data to identify TIME-related genes. To identify prognostic genes, we performed univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses. Based on these genes, we constructed a prognostic signature and delineated risk groups. To validate the prognostic signature, external validation was conducted. RESULTS CD8 + T cell infiltration was strongly correlated with glioma patient prognosis. We identified 115 CD8 + T cell-related genes through integrative analysis of bulk-seq data. CDCA5, KIF11, and KIF4A were found to be significant immune-related genes (IRGs) associated with overall survival in glioma patients and served as independent prognostic factors. We developed a prognostic nomogram that incorporated these genes, age, gender, and grade, providing a reliable tool for clinicians to predict patient survival probabilities. The nomogram's predictions were supported by calibration plots, further validating its accuracy. CONCLUSION In conclusion, our study identifies CD8 + T cell infiltration as a strong predictor of glioma patient outcomes and highlights the prognostic value of genes. The developed prognostic nomogram, incorporating these genes along with clinical factors, provides a reliable tool for predicting patient survival probabilities and has important implications for personalized treatment decisions in glioma.
Collapse
Affiliation(s)
- Xiaofang Lin
- Laboratory Department of Zengcheng Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianqiang Liu
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ni Zhang
- Department of Physical Medicine and Rehabilitation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Dexiang Zhou
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Yakang Liu
- Department of Physical Medicine and Rehabilitation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, 106 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Fang C, Han W, Tang C, Shen J, Ni B. Identification of RTP4 facilitating ovarian cancer by bioinformatics analysis and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03421-z. [PMID: 39249504 DOI: 10.1007/s00210-024-03421-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024]
Abstract
Ovarian cancer (OV) is the most malignant gynecological tumor in women, with poor prognosis and high mortality rate. This study aims to identify hub genes in OV and explore the role of Receptor transporter 4 (RTP4) in OV progression. Common differentially expressed genes (DEGs) were screened from two microarray datasets. GO and KEGG enrichment analysis were performed. Protein-protein interaction (PPI) network was constructed by STING. Kaplan-Meier plotter was used to analyze prognosis. The effect of target gene on immune infiltration was analyzed by TIMER. The proliferation, migration, and invasion of OV cells were measured by CCK-8, wound healing assay, and trans-well assay, respectively. A total of 293 common DEGs were selected from GSE12470 and GSE16709 datasets. Hub genes, EPCAM, KIFC1, RTP4, TAGLN, and ZFP36 were selected by PPI network. Kaplan-Meier plotter demonstrated that high expression of RTP4 was related to low overall survival in OV patients. The TIMER result showed that high expression of RTP4 promoted immune infiltration of CD8+ T cells, B cells, neutrophils, and dendritic cells in OV. Moreover, silencing RTP4 significantly inhibited the proliferation, migration, and invasion of OV cells. RTP4 was associated with the poor prognosis in OV. In summary, silencing RTP4 inhibited the proliferation, migration, and invasion of OV cells, having the potential to be a novel therapeutic target for OV.
Collapse
Affiliation(s)
- Chunyun Fang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Wenling Han
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Chen Tang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Junjing Shen
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, 341000, Jiangxi Province, China
| | - Bin Ni
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China.
| |
Collapse
|
5
|
Li Y, Cui Y, Wang Z, Wang L, Yu Y, Xiong Y. Development and validation of a hypoxia- and mitochondrial dysfunction- related prognostic model based on integrated single-cell and bulk RNA sequencing analyses in gastric cancer. Front Immunol 2024; 15:1419133. [PMID: 39165353 PMCID: PMC11333257 DOI: 10.3389/fimmu.2024.1419133] [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: 04/17/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction Gastric cancer (GC) remains a major global health threat ranking as the fifth most prevalent cancer. Hypoxia, a characteristic feature of solid tumors, significantly contributes to the malignant progression of GC. Mitochondria are the major target of hypoxic injury that promotes mitochondrial dysfunction during the development of cancers including GC. However, the gene signature and prognostic model based on hypoxia- and mitochondrial dysfunction-related genes (HMDRGs) in the prediction of GC prognosis have not yet been established. Methods The gene expression profile datasets of stomach cancer patients were retrieved from The Cancer Genome Atlas and the Gene Expression Omnibus databases. Prognostic genes were selected using Least Absolute Shrinkage and Selection Operator Cox (LASSO-Cox) regression analysis to construct a prognostic model. Immune infiltration was evaluated through ESTIMATE, CIBERSORT, and ssGSEA analyses. Tumor immune dysfunction and exclusion (TIDE) and immunophenoscore (IPS) were utilized to explore implications for immunotherapy. Furthermore, in vitro experiments were conducted to validate the functional roles of HMDRGs in GC cell malignancy. Results In this study, five HMDRGs (ZFP36, SERPINE1, DUSP1, CAV1, and AKAP12) were identified for developing a prognostic model in GC. This model stratifies GC patients into high- and low-risk groups based on median risk scores. A nomogram predicting overall survival (OS) was constructed and showed consistent results with observed OS. Immune infiltration analysis indicated that individuals in the high-risk group tend to exhibit increased immune cell infiltration. Additionally, analysis of cancer immunotherapy responses revealed that high-risk group patients exhibit poorer responses to cancer immunotherapy compared to the low-risk group. Immunohistochemistry (IHC) staining indicated that the expression levels of HMDRGs were remarkably correlated with GC, of which, SERPINE1 displayed the most pronounced up-regulation, while ZFP36 exhibited the most notable down-regulation in GC patients. Furthermore, in vitro investigation validated that SERPINE1 and ZFP36 contribute to the malignant processes of GC cells correlated with mitochondrial dysfunction. Conclusions This study presents a novel and efficient approach to evaluate GC prognosis and immunotherapy efficacy, and also provides insights into understanding the pathogenesis of GC.
Collapse
Affiliation(s)
- Yirong Li
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yue Cui
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Zhen Wang
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Liwei Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Yi Yu
- School of Medicine, Northwest University, Xi’an, Shaanxi, China
| | - Yuyan Xiong
- Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
6
|
Butt DQ, Harun MH, Che Jalil NA, Shamsuddin SH, Jaafar S, Ahmad B. Protumorigenic Interferon-Stimulated Genes in Cancer: A Comprehensive Review. Cureus 2024; 16:e63216. [PMID: 39070493 PMCID: PMC11279184 DOI: 10.7759/cureus.63216] [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] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Interferon-stimulated genes (ISGs), whose production is triggered by interferons, are known to defend the host from pathogenic and cancer-specific antigens, one of which is by inducing apoptosis in infected or mutated cells. It has been reported recently that specific ISGs aid cancer cells in evading immunosurveillance and inflammatory cells by inhibiting the apoptosis process. This report reviewed four apoptosis-regulating ISG proteins: interferon-stimulated gene 15 (ISG15), interferon alpha-inducible protein 27 (IFI27), interferon alpha-inducible protein 6 (IFI6), and radical S-adenosyl methionine domain containing 2 (RSAD2), demonstrating anti-apoptosis function, and considered them protumorigenic.
Collapse
Affiliation(s)
- Danial Qasim Butt
- Oral Medicine and Oral Pathology Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| | - Masitah Hayati Harun
- Oral Medicine and Oral Pathology Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| | - Nur Asyilla Che Jalil
- Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| | | | - Saidi Jaafar
- Basic Sciences Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| | - Basaruddin Ahmad
- Biostatistics Unit, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, MYS
| |
Collapse
|
7
|
Mathew Thomas V, Chigarira B, Gebrael G, Sayegh N, Tripathi N, Nussenzveig R, Jo Y, Dal E, Galarza Fortuna G, Li H, Sahu KK, Srivastava A, Maughan BL, Agarwal N, Swami U. Differential Tumor Gene Expression Profiling of Patients With Prostate Adenocarcinoma on the Basis of BMI. JCO Precis Oncol 2024; 8:e2300574. [PMID: 38781543 DOI: 10.1200/po.23.00574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/23/2023] [Accepted: 03/18/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE An increased BMI is linked to increased prostate adenocarcinoma incidence and mortality. Baseline tumor gene expression profiling (GEP) can provide a comprehensive picture of the biological processes related to treatment response and disease progression. We interrogate and validate the underlying differences in tumor GEP on the basis of BMI in patients with prostate adenocarcinoma. METHODS The inclusion criteria consisted of histologically confirmed prostate adenocarcinoma and the availability of RNA sequencing data obtained from treatment-naïve primary prostate tissue. RNA sequencing was performed by a Clinical Laboratory Improvement Amendments-certified laboratory (Tempus or Caris Life Sciences). The Tempus cohort was used for interrogation and the Caris cohort for validation. Patients were stratified on the basis of BMI at the time of prostate cancer diagnosis: BMI-high (BMIH; BMI ≥30) and BMI-low (BMIL; BMI <30). Differential gene expression analysis between the two cohorts was conducted using the DEseq2 pipeline. The resulting GEPs were further analyzed using Gene Set Enrichment software to identify pathways that exhibited enrichment in each cohort. RESULTS Overall, 102 patients were eligible, with 60 patients in the Tempus cohort (BMIL = 38, BMIH = 22) and 42 patients in the Caris cohort (BMIL = 24, BMIH = 18). Tumor tissues obtained from patients in the BMIL group exhibited higher expression of genes associated with inflammation pathways. BMIH displayed increased expression of genes involved in pathways such as heme metabolism and androgen response. CONCLUSION Our study shows the upregulation of distinct genomic pathways in BMIL compared with BMIH patients with prostate cancer. These hypothesis-generating data could explain different survival outcomes in both groups and guide personalized therapy for men with prostate cancer.
Collapse
Affiliation(s)
- Vinay Mathew Thomas
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Beverly Chigarira
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nishita Tripathi
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Roberto Nussenzveig
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Yeonjung Jo
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT
| | - Emre Dal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Gliceida Galarza Fortuna
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Haoran Li
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Cancer Center, Westwood, KS
| | - Kamal Kant Sahu
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Ayana Srivastava
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Benjamin L Maughan
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
8
|
Mao J, Chen L, Qian S, Wang Y, Zhao B, Zhao Q, Lu B, Mao X, Zhai P, Zhang Y, Zhang L, Sun X. Transcriptome network analysis of inflammation and fibrosis in keloids. J Dermatol Sci 2024; 113:62-73. [PMID: 38242738 DOI: 10.1016/j.jdermsci.2023.12.007] [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: 03/30/2023] [Revised: 11/07/2023] [Accepted: 12/24/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND Keloid (KL) is a common benign skin tumor. KL is typically characterized by significant fibrosis and an intensive inflammatory response. Therefore, a comprehensive understanding of the interactions between cellular inflammation and fibrotic cells is essential to elucidate the mechanisms driving the progression of KL and to develop therapeutics. OBJECTIVE Investigate the transcriptome landscape of inflammation and fibrosis in keloid scars. METHODS In this paper, we performed transcriptome sequencing and microRNA (miRNA) sequencing on unselected live cells from six human keloid tissues and normal skin tissues to elucidate a comprehensive transcriptome landscape. In addition, we used single-cell RNA sequencing (scRNA-seq) analysis to analyze intercellular communication networks and enrich fibroblast populations in two additional keloid and normal skin samples to study fibroblast diversity. RESULTS By RNA sequencing and a miRNA-mRNA-PPI network analysis, we identified miR-615-5p and miR-122b-3p as possible miRNAs associated with keloids, as they differed most significantly in keloids. Similarly, COL3A1, COL1A2, THBS2, TNC, IGTA, THBS4, TGFB3 as genes with significant differences in keloid may be associated with keloid development. Using single-cell RNA sequencing data from 24,086 cells collected from normal or keloid, we report reconstructed intercellular signaling network analysis and aggregation to modules associated with specific cell subpopulations at the cellular level for keloid alterations. CONCLUSIONS Our multitranscriptomic dataset delineates inflammatory and fibro heterogeneity of human keloids, underlining the importance of intercellular crosstalk between inflammatory cells and fibro cells and revealing potential therapeutic targets.
Collapse
Affiliation(s)
- Jiayi Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Chen
- Department of Plastic and Burn Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shutong Qian
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhuan Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Binfan Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyu Zhao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bolun Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiyuan Mao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peisong Zhai
- Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuguang Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Liucheng Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiaoming Sun
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Silvestri R, Nicolì V, Gangadharannambiar P, Crea F, Bootman MD. Calcium signalling pathways in prostate cancer initiation and progression. Nat Rev Urol 2023; 20:524-543. [PMID: 36964408 DOI: 10.1038/s41585-023-00738-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/26/2023]
Abstract
Cancer cells proliferate, differentiate and migrate by repurposing physiological signalling mechanisms. In particular, altered calcium signalling is emerging as one of the most widespread adaptations in cancer cells. Remodelling of calcium signalling promotes the development of several malignancies, including prostate cancer. Gene expression data from in vitro, in vivo and bioinformatics studies using patient samples and xenografts have shown considerable changes in the expression of various components of the calcium signalling toolkit during the development of prostate cancer. Moreover, preclinical and clinical evidence suggests that altered calcium signalling is a crucial component of the molecular re-programming that drives prostate cancer progression. Evidence points to calcium signalling re-modelling, commonly involving crosstalk between calcium and other cellular signalling pathways, underpinning the onset and temporal progression of this disease. Discrete alterations in calcium signalling have been implicated in hormone-sensitive, castration-resistant and aggressive variant forms of prostate cancer. Hence, modulation of calcium signals and downstream effector molecules is a plausible therapeutic strategy for both early and late stages of prostate cancer. Based on this premise, clinical trials have been undertaken to establish the feasibility of targeting calcium signalling specifically for prostate cancer.
Collapse
Affiliation(s)
| | - Vanessa Nicolì
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | | | - Francesco Crea
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Martin D Bootman
- Cancer Research Group, School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
10
|
Yao K, Zhang R, Li L, Liu M, Feng S, Yan H, Zhang Z, Xie D. The signature of cuproptosis-related immune genes predicts the tumor microenvironment and prognosis of prostate adenocarcinoma. Front Immunol 2023; 14:1181370. [PMID: 37600770 PMCID: PMC10433769 DOI: 10.3389/fimmu.2023.1181370] [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: 03/07/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Cuproptosis plays a crucial role in cancer, and different subtypes of cuproptosis have different immune profiles in prostate adenocarcinoma (PRAD). This study aimed to investigate immune genes associated with cuproptosis and develop a risk model to predict prognostic characteristics and chemotherapy/immunotherapy responses of patients with PRAD. Methods The CIBERSORT algorithm was used to evaluate the immune and stromal scores of patients with PRAD in The Cancer Genome Atlas (TCGA) cohort. Validation of differentially expressed genes DLAT and DLD in benign and malignant tissues by immunohistochemistry, and the immune-related genes of DLAT and DLD were further screened. Univariable Cox regression were performed to select key genes. Least absolute shrinkage and selection operator (LASSO)-Cox regression analyse was used to develop a risk model based on the selected genes. The model was validated in the TCGA, Memorial Sloan-Kettering Cancer Center (MSKCC) and Gene Expression Omnibus (GEO) datasets, as well as in this study unit cohort. The genes were examined via functional enrichment analysis, and the tumor immune features, tumor mutation features and copy number variations (CNVs) of patients with different risk scores were analysed. The response of patients to multiple chemotherapeutic/targeted drugs was assessed using the pRRophetic algorithm, and immunotherapy was inferred by the Tumor Immune Dysfunction and Exclusion (TIDE) and immunophenoscore (IPS). Results Cuproptosis-related immune risk scores (CRIRSs) were developed based on PRLR, DES and LECT2. High CRIRSs indicated poor overall survival (OS), disease-free survival (DFS) in the TCGA-PRAD, MSKCC and GEO datasets and higher T stage and Gleason scores in TCGA-PRAD. Similarly, in the sample collected by the study unit, patients with high CRIRS had higher T-stage and Gleason scores. Additionally, higher CRIRSs were negatively correlated with the abundance of activated B cells, activated CD8+ T cells and other stromal or immune cells. The expression of some immune checkpoints was negatively correlated with CRIRSs. Tumor mutational burden (TMB), mutant-allele tumor heterogeneity (MATH) and copy number variation (CNV) scores were all higher in the high-CRIRS group. Multiple chemotherapeutic/targeted drugs and immunotherapy had better responsiveness in the low-CRIRS group. Conclusion Overall, lower CRIRS indicated better response to treatment strategies and better prognostic outcomes.
Collapse
Affiliation(s)
- Kai Yao
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rumeng Zhang
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Liang Li
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingdong Liu
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shiyao Feng
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haixin Yan
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihui Zhang
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dongdong Xie
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Urology, Affiliated Fuyang Hospital of Anhui Medical University, Fuyang, Anhui, China
| |
Collapse
|
11
|
Chaddad A, Tan G, Liang X, Hassan L, Rathore S, Desrosiers C, Katib Y, Niazi T. Advancements in MRI-Based Radiomics and Artificial Intelligence for Prostate Cancer: A Comprehensive Review and Future Prospects. Cancers (Basel) 2023; 15:3839. [PMID: 37568655 PMCID: PMC10416937 DOI: 10.3390/cancers15153839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The use of multiparametric magnetic resonance imaging (mpMRI) has become a common technique used in guiding biopsy and developing treatment plans for prostate lesions. While this technique is effective, non-invasive methods such as radiomics have gained popularity for extracting imaging features to develop predictive models for clinical tasks. The aim is to minimize invasive processes for improved management of prostate cancer (PCa). This study reviews recent research progress in MRI-based radiomics for PCa, including the radiomics pipeline and potential factors affecting personalized diagnosis. The integration of artificial intelligence (AI) with medical imaging is also discussed, in line with the development trend of radiogenomics and multi-omics. The survey highlights the need for more data from multiple institutions to avoid bias and generalize the predictive model. The AI-based radiomics model is considered a promising clinical tool with good prospects for application.
Collapse
Affiliation(s)
- Ahmad Chaddad
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
- The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
| | - Guina Tan
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | - Xiaojuan Liang
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | - Lama Hassan
- School of Artificial Intelligence, Guilin Universiy of Electronic Technology, Guilin 541004, China
| | | | - Christian Desrosiers
- The Laboratory for Imagery, Vision and Artificial Intelligence, École de Technologie Supérieure (ETS), Montreal, QC H3C 1K3, Canada
| | - Yousef Katib
- Department of Radiology, Taibah University, Al Madinah 42361, Saudi Arabia
| | - Tamim Niazi
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
12
|
Liu S, Li F, Ma Q, Du M, Wang H, Zhu Y, Deng L, Gao W, Wang C, Liu Y, Zhao Z, Liu H, Wang R, Tian Y, Hu M, Wan Y, Lu W, Zhang M, Zhao M, Cao Y, Zhang H, Wang W, Wang H, Wang Y. OX40L-Armed Oncolytic Virus Boosts T-cell Response and Remodels Tumor Microenvironment for Pancreatic Cancer Treatment. Theranostics 2023; 13:4016-4029. [PMID: 37554264 PMCID: PMC10405835 DOI: 10.7150/thno.83495] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/27/2023] [Indexed: 08/10/2023] Open
Abstract
Rationale: The resistance of pancreatic ductal adenocarcinoma (PDAC) to immunotherapies is caused by the immunosuppressive tumor microenvironment (TME) and dense extracellular matrix. Currently, the efficacy of an isolated strategy targeting stromal desmoplasia or immune cells has been met with limited success in the treatment of pancreatic cancer. Oncolytic virus (OV) therapy can remodel the TME and damage tumor cells either by directly killing them or by enhancing the anti-tumor immune response, which holds promise for the treatment of PDAC. This study aimed to investigate the therapeutic effect of OX40L-armed OV on PDAC and to elucidate the underlying mechanisms. Methods: Murine OX40L was inserted into herpes simplex virus-1 (HSV-1) to construct OV-mOX40L. Its expression and function were assessed using reporter cells, cytopathic effect, and immunogenic cell death assays. The efficacy of OV-mOX40L was then evaluated in a KPC syngeneic mouse model. Tumor-infiltrating immune and stromal cells were analyzed using flow cytometry and single-cell RNA sequencing to gain insight into the mechanisms of oncolytic virotherapy. Results: OV-mOX40L treatment delayed tumor growth in KPC tumor-bearing C57BL/6 mice. It also boosted the tumor-infiltrating CD4+ T cell response, mitigated cytotoxic T lymphocyte (CTL) exhaustion, and reduced the number of regulatory T cells. The treatment of OV-mOX40L reprogrammed macrophages and neutrophils to a more pro-inflammatory anti-tumor state. In addition, the number of myofibroblastic cancer-associated fibroblasts (CAF) was reduced after treatment. Based on single-cell sequencing analysis, OV-mOX40L, in combination with anti-IL6 and anti-PD-1, significantly extended the lifespan of PDAC mice. Conclusion: OV-mOX40L converted the immunosuppressive tumor immune microenvironment to a more activated state, remodeled the stromal matrix, and enhanced T cell response. OV-mOX40L significantly prolonged the survival of PDAC mice, either as a monotherapy or in combination with synergistic antibodies. Thus, this study provides a multimodal therapeutic strategy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Shiyu Liu
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Fan Li
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Qiongqiong Ma
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Mingjuan Du
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Haoran Wang
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
| | - Yiping Zhu
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Li Deng
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Wenrui Gao
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Chunlei Wang
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Yanqin Liu
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Zhuoqian Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
| | - Huanzhen Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Ruikun Wang
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
| | - Yujie Tian
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Manli Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Yajuan Wan
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Meng Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Youjia Cao
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Wei Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Hui Wang
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| | - Yuan Wang
- State Key Laboratory of Medicinal Chemical Biology and College of life science, Nankai University, Tianjin, China
- CNBG-Nankai University Joint Research and Development Center, Tianjin, China
| |
Collapse
|
13
|
Qu T, Zhang W, Yan C, Ren D, Wang Y, Guo Y, Guo Q, Wang J, Liu L, Han L, Li L, Huang Q, Cao L, Ye Z, Zhang B, Zhao Q, Cao W. ISG15 targets glycosylated PD-L1 and promotes its degradation to enhance antitumor immune effects in lung adenocarcinoma. J Transl Med 2023; 21:341. [PMID: 37217923 DOI: 10.1186/s12967-023-04135-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/16/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Immunocheckpoint inhibitors (ICIs) have been widely used in the clinical treatment of lung cancer. Although clinical studies and trials have shown that patients can benefit significantly after PD-1/PD-L1 blocking therapy, less than 20% of patients can benefit from ICIs therapy due to tumor heterogeneity and the complexity of immune microenvironment. Several recent studies have explored the immunosuppression of PD-L1 expression and activity by post-translational regulation. Our published articles demonstrate that ISG15 inhibits lung adenocarcinoma progression. Whether ISG15 can enhance the efficacy of ICIs by modulating PD-L1 remains unknown. METHODS The relationship between ISG15 and lymphocyte infiltration was identified by IHC. The effects of ISG15 on tumor cells and T lymphocytes were assessed using RT-qPCR and Western Blot and in vivo experiments. The underlying mechanism of PD-L1 post-translational modification by ISG15 was revealed by Western blot, RT-qPCR, flow cytometry, and Co-IP. Finally, we performed validation in C57 mice as well as in lung adenocarcinoma tissues. RESULTS ISG15 promotes the infiltration of CD4+ T lymphocytes. In vivo and in vitro experiments demonstrated that ISG15 induces CD4+ T cell proliferation and invalidity and immune responses against tumors. Mechanistically, we demonstrated that the ubiquitination-like modifying effect of ISG15 on PD-L1 increased the modification of K48-linked ubiquitin chains thus increasing the degradation rate of glycosylated PD-L1 targeting proteasomal pathway. The expression of ISG15 and PD-L1 was negatively correlated in NSCLC tissues. In addition, reduced accumulation of PD-L1 by ISG15 in mice also increased splenic lymphocyte infiltration as well as promoted cytotoxic T cell infiltration in the tumor microenvironment, thereby enhancing anti-tumor immunity. CONCLUSIONS The ubiquitination modification of PD-L1 by ISG15 increases K48-linked ubiquitin chain modification, thereby increasing the degradation rate of glycosylated PD-L1-targeted proteasome pathway. More importantly, ISG15 enhanced the sensitivity to immunosuppressive therapy. Our study shows that ISG15, as a post-translational modifier of PD-L1, reduces the stability of PD-L1 and may be a potential therapeutic target for cancer immunotherapy.
Collapse
Affiliation(s)
- Tongyuan Qu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenshuai Zhang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chenhui Yan
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Danyang Ren
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yalei Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yuhong Guo
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qianru Guo
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinpeng Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Liren Liu
- Department of Gastrointestinal Cancer Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lei Han
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lingmei Li
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qiujuan Huang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Lu Cao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bin Zhang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerKey Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of EducationTianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Wenfeng Cao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
14
|
Nguyen HM, Gaikwad S, Oladejo M, Agrawal MY, Srivastava SK, Wood LM. Interferon stimulated gene 15 (ISG15) in cancer: An update. Cancer Lett 2023; 556:216080. [PMID: 36736853 DOI: 10.1016/j.canlet.2023.216080] [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: 11/16/2022] [Revised: 01/17/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023]
Abstract
Among the plethora of defense mechanisms which a host elicits after pathogen invasion, type 1 interferons play a central role in regulating the immune system's response. They induce several interferon-stimulated genes (ISGs) which play a diverse role once activated. Over the past few decades, there have been several studies exploring the role of ISGs in cancer and ISG15 is among the most studied for its pro and anti-tumorigenic role. In this review, we aim to provide an update on the recent observations and findings related to ISG15 in cancer. We provide a brief overview about the initial observations and important historical findings which helped scientists understand structure and function of ISG15. We aim to provide an overview of ISG15 in cancer with an emphasis on studies which delve into the molecular mechanism of ISG15 in modulating the tumor microenvironment. Further, the dysregulation of ISG15 in cancer and the molecular mechanisms associated with its pro and anti-tumor roles are discussed in respective cancer types. Finally, we discuss multiple therapeutic applications of ISG15 in current cancer therapy.
Collapse
Affiliation(s)
- Hong-My Nguyen
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Shreyas Gaikwad
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Manas Yogendra Agrawal
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA
| | - Laurence M Wood
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Abilene, TX, 79601, USA; Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, USA.
| |
Collapse
|
15
|
Angel CZ, Stafford MYC, McNally CJ, Nesbitt H, McKenna DJ. MiR-21 Is Induced by Hypoxia and Down-Regulates RHOB in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15041291. [PMID: 36831632 PMCID: PMC9954526 DOI: 10.3390/cancers15041291] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Tumour hypoxia is a well-established contributor to prostate cancer progression and is also known to alter the expression of several microRNAs. The over-expression of microRNA-21 (miR-21) has been consistently linked with many cancers, but its role in the hypoxic prostate tumour environment has not been well studied. In this paper, the link between hypoxia and miR-21 in prostate cancer is investigated. A bioinformatic analysis of The Cancer Genome Atlas (TCGA) prostate biopsy datasets shows the up-regulation of miR-21 is significantly associated with prostate cancer and clinical markers of disease progression. This up-regulation of miR-21 expression was shown to be caused by hypoxia in the LNCaP prostate cancer cell line in vitro and in an in vivo prostate tumour xenograft model. A functional enrichment analysis also revealed a significant association of miR-21 and its target genes with processes related to cellular hypoxia. The over-expression of miR-21 increased the migration and colony-forming ability of RWPE-1 normal prostate cells. In vitro and in silico analyses demonstrated that miR-21 down-regulates the tumour suppressor gene Ras Homolog Family Member B (RHOB) in prostate cancer. Further a TCGA analysis illustrated that miR-21 can distinguish between different patient outcomes following therapy. This study presents evidence that hypoxia is a key contributor to the over-expression of miR-21 in prostate tumours, which can subsequently promote prostate cancer progression by suppressing RHOB expression. We propose that miR-21 has good potential as a clinically useful diagnostic and prognostic biomarker of hypoxia and prostate cancer.
Collapse
Affiliation(s)
- Charlotte Zoe Angel
- Genomic Medicine Research Group, Ulster University, Cromore Road, Coleraine BT52 1SA, UK
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK
| | | | - Christopher J. McNally
- Genomic Medicine Research Group, Ulster University, Cromore Road, Coleraine BT52 1SA, UK
| | - Heather Nesbitt
- Genomic Medicine Research Group, Ulster University, Cromore Road, Coleraine BT52 1SA, UK
| | - Declan J. McKenna
- Genomic Medicine Research Group, Ulster University, Cromore Road, Coleraine BT52 1SA, UK
- Correspondence:
| |
Collapse
|
16
|
Peng C, Ye H, li Z, Duan X, Yang W, Yi Z. Multi-omics characterization of a scoring system to quantify hypoxia patterns in patients with head and neck squamous cell carcinoma. J Transl Med 2023; 21:15. [PMID: 36627705 PMCID: PMC9830846 DOI: 10.1186/s12967-022-03869-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The 5-year survival rate of patients with head and neck squamous cell carcinoma (HNSCC) remains < 50%. Hypoxia patterns are a hallmark of HNSCC that are associated with its occurrence and progression. However, the precise role of hypoxia during HNSCC, such as the relationship between hypoxia, tumor immune landscape and cell communication orchestration remains largely unknown. The current study integrated data from bulk and single-cell RNA sequencing analyses to define the relationship between hypoxia and HNSCC. METHODS A scoring system named the hypoxia score (HS) was constructed based on hypoxia-related genes (HRGs) expression. The predictive value of HS response for patient outcomes and different treatments was evaluated. Single-cell datasets and cell communication were utilized to rule out cell populations which hypoxia targeted on. RESULTS The survival outcomes, immune/Estimate scores, responses to targeted inhibitors, and chemotherapeutic, and immunotherapy responses were distinct between a high HS group and a low HS group (all P < 0.05). Single-cell datasets showed different distributions of HS in immune cell populations (P < 0.05). Furthermore, HLA-DPA1/CD4 axis was identified as a unique interaction between CD4 + T Conv and pDC cells. CONCLUSIONS Altogether, the quantification for hypoxia patterns is a potential biomarker for prognosis, individualized chemotherapeutic and immunotherapy strategies. The portrait of cell communication characteristics over the HNSCC ecosystem enhances the understanding of hypoxia patterns in HNSCC.
Collapse
Affiliation(s)
- Cong Peng
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Huiping Ye
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Zhengyang li
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiaofeng Duan
- grid.459540.90000 0004 1791 4503Department of Oral and Maxillofacial Surgery, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Wen Yang
- grid.452244.1Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhuguang Yi
- grid.459540.90000 0004 1791 4503Department of Otolaryngology, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
17
|
Man YG, Mannion C, Jewett A, Hsiao YH, Liu A, Semczuk A, Zarogoulidis P, Gapeev AB, Cimadamore A, Lee P, Lopez-Beltran A, Montironi R, Massari F, Lu X, Cheng L. The most effective but largely ignored target for prostate cancer early detection and intervention. J Cancer 2022; 13:3463-3475. [PMID: 36313040 PMCID: PMC9608211 DOI: 10.7150/jca.72973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 10/01/2022] [Indexed: 11/05/2022] Open
Abstract
Over the past two decades, the global efforts for the early detection and intervention of prostate cancer seem to have made significant progresses in the basic researches, but the clinic outcomes have been disappointing: (1) prostate cancer is still the most common non-cutaneous cancer in Europe in men, (2) the age-standardized prostate cancer rate has increased in nearly all Asian and African countries, (3) the proportion of advanced cancers at the diagnosis has increased to 8.2% from 3.9% in the USA, (4) the worldwide use of PSA testing and digital rectal examination have failed to reduce the prostate cancer mortality, and (5) there is still no effective preventive method to significantly reduce the development, invasion, and metastasis of prostate cancer… Together, these facts strongly suggest that the global efforts during the past appear to be not in a correlated target with markedly inconsistent basic research and clinic outcomes. The most likely cause for the inconsistence appears due to the fact that basic scientific studies are traditionally conducted on the cell lines and animal models, where it is impossible to completely reflect or replicate the in vivo status. Thus, we would like to propose the human prostate basal cell layer (PBCL) as “the most effective target for the early detection and intervention of prostate cancer”. Our proposal is based on the morphologic, immunohistochemical and molecular evidence from our recent studies of normal and cancerous human prostate tissues with detailed clinic follow-up data. We believe that the human tissue-derived basic research data may provide a more realistic roadmap to guide the clinic practice and to avoid the potential misleading from in vitro and animal studies.
Collapse
Affiliation(s)
- Yan-gao Man
- Department of Pathology, Hackensack Meridian School of Medicine, Nutley, NJ, USA,✉ Corresponding authors: Yan-gao Man., MD., PhD. E-mail: or or Liang Cheng., MD. E-mail: or
| | - Ciaran Mannion
- Department of Pathology, Hackensack Meridian School of Medicine, Nutley, NJ, USA
| | - Anahid Jewett
- Tumor Immunology Laboratory, Jonsson Comprehensive Cancer Center, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA
| | - Yi-Hsuan Hsiao
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua, Taiwan
| | - Aijun Liu
- Department of Pathology, Chinese PLA General Hospital 7 th Medical Center, Beijing, China
| | - Andrzej Semczuk
- II ND Department of Gynecology, Lublin Medical University, Lublin, Poland
| | - Paul Zarogoulidis
- Pulmonary-Oncology Department, "Theageneio" Cancer Hospital, Thessaloniki, Greece
| | - Andrei B. Gapeev
- Laboratory of Biological Effects of Non-Ionizing Radiation, Institute of Cell Biophysics, Russian Academy of Sciences, Russian Federation
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Peng Lee
- Department of Pathology, New York University School of Medicine, New York, NY, USA.,Department of Pathology, New York Harbor Healthcare System, New York, NY, USA
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, Cordoba University Medical School, Cordoba, Spain
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Department of Clinical & Molecular Sciences, Polytechnic University of the Marche Region, Ancona, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, USA.,Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Brown University Medical School
- Lifespan Academic Medical Center, RI, USA.,✉ Corresponding authors: Yan-gao Man., MD., PhD. E-mail: or or Liang Cheng., MD. E-mail: or
| |
Collapse
|
18
|
Integrative Analysis of Bulk RNA-Seq and Single-Cell RNA-Seq Unveils the Characteristics of the Immune Microenvironment and Prognosis Signature in Prostate Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6768139. [PMID: 35909899 PMCID: PMC9325591 DOI: 10.1155/2022/6768139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/21/2022] [Indexed: 12/01/2022]
Abstract
The immune microenvironment is a culmination of the collaborative effort of immune cells and is important in cancer development. The underlying mechanisms of the tumor immune microenvironment in regulating prostate cancer (PRAD) are unclear. In the current study, 144 natural killer cell-related genes were identified using differential expression, single-sample gene set enrichment analysis, and weighted gene coexpression network analysis. Furthermore, VCL, ACTA2, MYL9, MYLK, MYH11, TPM1, ACTG2, TAGLN, and FLNC were selected as hub genes via the protein-protein interaction network. Based on the expression patterns of the hub genes, endothelial, epithelial, and tissue stem cells were identified as key cell subpopulations, which could regulate PRAD via immune response, extracellular signaling, and protein formation. Moreover, 27 genes were identified as prognostic signatures and used to construct the risk score model. Receiver operating characteristic curves revealed the good performance of the risk score model in both the training and testing datasets. Different chemotherapeutic responses were observed between the low- and high-risk groups. Additionally, a nomogram based on the risk score and other clinical features was established to predict the 1-, 3-, and 5-year progression-free interval of patients with PRAD. This study provides novel insights into the molecular mechanisms of the immune microenvironment and its role in the pathogenesis of PARD. The identification of key cell subpopulations has a potential therapeutic and prognostic use in PRAD.
Collapse
|