1
|
Artamonova N, Kafka M, Faiss L, Avetisyan D, Puche Sanz I, La Bombarda G, Iacono G, Zattoni F, Steiner E, D’Elia C, Pycha A, Ladurner M, Jagodic S, Gandaglia G, Heidegger I. Impact of Renin-Angiotensin System Inhibitors on Disease Characteristics in Patients with Localized Prostate Cancer Treated with Radical Prostatectomy: A European Association of Urology Young Academic Urologists Prostate Cancer Working Group Multi-institutional Study. EUR UROL SUPPL 2024; 69:105-111. [PMID: 39430410 PMCID: PMC11490865 DOI: 10.1016/j.euros.2024.09.005] [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: 09/14/2024] [Indexed: 10/22/2024] Open
Abstract
Background and objective Collagen biosynthesis is intricately involved in the development and progression of solid tumors. Renin-angiotensin system inhibitors (RASi) impede TGF-β-mediated collagen synthesis in tumors by hindering activation of the angiotensin receptor. Our aim was to investigate a potential association between RASi use and the aggressiveness of prostate cancer (PCa). Methods We conducted a retrospective multicenter analysis for a cohort of 1250 patients with PCa who underwent radical prostatectomy (RP) between 1990 and 2023 in four European high-volume centers. The study cohort comprised 625 RASi-treated patients and 625 age-matched RASi-naïve patients. Data for various parameters were collected, including age at RP, body mass index (BMI), prostate volume, prostate-specific antigen (PSA), percentage of free PSA, Gleason score (GS) at biopsy and RP, TNM stage, and the rate of biochemical recurrence (BCR). Clinical parameters for patients with and without RASi treatment were documented. Differences between the groups were compared using a Mann-Whitney U test and χ2 tests. Survival analyses were performed using the Kaplan-Meier method. Key findings and limitations As expected, the RASi group had higher BMI levels than the RASi-naïve group (p < 0.001). However, RASi use was not associated with key markers of PCa aggressiveness such as GS upgrading from biopsy to RP (p = 0.089), surgical margin status (p = 0.109), and lymph node involvement (p = 0.33). Moreover, there were no significant differences between the groups in BCR incidence (p = 0.258) or the time to BCR (p = 0.683). Conclusions and clinical implications Our findings indicate that RASi therapy does not have a significant effect on the biological aggressiveness of PCa. Patient summary We analyzed data for 1250 patients with prostate cancer and found that the use of a commonly prescribed high blood pressure medication was not associated with a less aggressive form of localized prostate cancer.
Collapse
Affiliation(s)
| | - Mona Kafka
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Laura Faiss
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - David Avetisyan
- UGC Urología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Ignacio Puche Sanz
- UGC Urología, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | - Gennaio Iacono
- Department of Urology, University of Padova, Padova, Italy
| | - Fabio Zattoni
- Department of Urology, University of Padova, Padova, Italy
- Department of Medicine, University of Padova, Padua, Italy
| | - Eberhard Steiner
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Caroline D’Elia
- Department of Urology, Zentralkankenhaus Bozen, Bozen, Italy
| | - Armin Pycha
- Department of Urology, Zentralkankenhaus Bozen, Bozen, Italy
| | - Michael Ladurner
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | - Samed Jagodic
- Department of Urology, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Giorgio Gandaglia
- Department of Urology, Urological Research Institute Vita-Salute University and San Raffaele Hospital, Milan, Italy
| | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
2
|
Zhang R, Yin H, Yang M, Liu J, Zhen D, Zhang Z. Advanced progress of the relationship between renin-angiotensin-aldosterone system inhibitors and cancers. J Hypertens 2024; 42:1862-1873. [PMID: 39248142 DOI: 10.1097/hjh.0000000000003836] [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: 03/08/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024]
Abstract
Hypertension and cancers are the most common causes of death in humans, as well as common co-diseases among elderly population. Studies have shown that hypertension is associated with carcinogenesis. The renin-angiotensin-aldosterone system (RAAS) is a crucial regulatory system of blood pressure, fluid, and electrolyte homeostasis, which plays an essential role in the pathogenesis of hypertension, whose mechanism is relatively clear. Studies have indicated that RAAS also widely exists in cancer tissues of different systems, which can affect the risk of cancers by stimulating cancer angiogenesis, participating in cancer-related oxidative stress, and regulating cancer-related immunity. Therefore, inhibiting RAAS activity seems beneficial to decreasing the risk of cancers. As one of the most commonly used antihypertensive drugs, RAAS inhibitors have been widely used in clinical practice. However, the conclusions of clinical studies on the relationship between RAAS inhibitors and cancers are not entirely consistent, which has been widely concerned by clinicians. The latest findings suggest that while RAAS inhibitors may reduce the risk of digestive cancers, respiratory cancers, urological cancers, gynecological cancers, and skin cancers, ACEIs may increase the risk of lung cancer, endometrial cancer, basal cell carcinoma, and squamous cell carcinoma. This article comprehensively reviews animal experiments, clinical studies, and meta-analyses on the relationship between RAAS inhibitors and cancers, to provide references for related studies in the future.
Collapse
Affiliation(s)
- Ruixing Zhang
- The First Clinical Medical College, Lanzhou University
- Department of Heart Center
| | - Hongtao Yin
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | - Mengdi Yang
- The First Clinical Medical College, Lanzhou University
| | - Jinjin Liu
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | - Donghu Zhen
- Department of Endocrinology, The First Hospital of Lanzhou University, Chengguan District, Lanzhou, Gansu, China
| | | |
Collapse
|
3
|
Zhou J, Zhang K, Yang T, Li A, Li M, Peng X, Chen M. Identification and validation of a glycosyltransferase gene signature as a novel prognostic model for lung adenocarcinoma. Heliyon 2024; 10:e29383. [PMID: 38644835 PMCID: PMC11033141 DOI: 10.1016/j.heliyon.2024.e29383] [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: 01/12/2024] [Revised: 04/07/2024] [Accepted: 04/07/2024] [Indexed: 04/23/2024] Open
Abstract
Background The role of glycosyltransferase (GT) genes in lung adenocarcinoma (LUAD) needs further elucidation. Thus, our study aims to identify the prognostic gene signature of LUAD and explore its molecular functions. Methods We initially extracted GT gene sets from the database, and obtained mRNA expression levels and clinical data from The Cancer Genome Atlas (TCGA) database. For constructing a prognostic model for GT genes, we utilized univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses. Using the model, patients were categorized into high- and low-risk groups. Additionally, we evaluated differences in tumor immune infiltration between these groups and identified potential therapeutic drugs. Finally, we experimentally validated the expression levels of these crucial prognostic genes. Results We developed a risk score comprising nine GT genes (C1GALT1, FUT1, GALNT2, PLOD2, POMK, PYGB, ST3GAL6, UGT2B11, UGT3A1). Patients were then categorized into low- and high-risk groups based on this score. The low-risk group showed superior overall survival (OS) compared to the high-risk group. There were significantly distinct tumor immune microenvironment statuses observed between the two groups. We identified potential therapeutic drugs, including the MEK inhibitor (PD-184352). Finally, we verified the expression of these nine GT genes through immunohistochemistry (IHC) staining and quantitative real-time PCR (qPCR). Conclusion We identified a distinct LUAD GT gene signature, and these differentially expressed mRNAs could serve as valuable prognostic biomarkers and therapeutic targets. Furthermore, we experimentally validated their expression levels and identified potential therapeutic agents.
Collapse
Affiliation(s)
| | | | - Tian Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Anqi Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Meng Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Xiaojing Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Mingwei Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| |
Collapse
|
4
|
Han WJ, He P. A novel tumor microenvironment-related gene signature with immune features for prognosis of lung squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:13137-13154. [PMID: 37479755 DOI: 10.1007/s00432-023-05042-0] [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: 05/13/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023]
Abstract
PURPOSE Lung squamous cell carcinoma (LUSC) is an aggressive subset of non-small-cell lung cancer (NSCLC). The tumor microenvironment (TME) plays an important role in the development of LUSC. We aim to identify potential therapeutic targets and a TME-related prognostic signature and for LUSC. METHODS TME-related genes were obtained from TCGA-LUSC dataset. LUSC samples were clustered by the non-negative matrix clustering algorithm (NMF). The prognostic signature was constructed through univariate Cox regression, multivariate Cox regression, and the least absolute shrinkage and selection operator (LASSO) analyses. Gene set enrichment analysis (GSEA) was carried out to explore the enrichment pathways. RESULTS This study constructed a prognostic signature which contained 12 genes: HHIPL2, PLK4, SLC6A4, LSM1, TSLP, P4HA1, AMH, CLDN5, NRTN, CDH2, PTGIS, and STX1A. Patients were classified into high-risk and low-risk groups according to the median risk score of this signature. Compared with low-risk group patients, patients in high-risk group patients had poorer overall survival, which demonstrated this signature was an independent prognostic factor. Besides, correlation analysis and GSEA results revealed that genes of this signature were correlated with immune cells and drug response. CONCLUSION Our novel signature based on 12 TME-related genes might be applied as an independent prognostic indicator. Importantly, the signature could be a promising biomarker and accurately predict the prognosis of LUSC patients.
Collapse
Affiliation(s)
- Wan Jia Han
- Beijing Normal University, Beijing, China.
- Sichuan Second Hospital of TCM, Chengdu, China.
| | - Pengzhi He
- Beijing Normal University, Beijing, China
- Sichuan Second Hospital of TCM, Chengdu, China
| |
Collapse
|
5
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [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: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
6
|
Xia J, Li S, Liu S, Zhang L. Aldehyde dehydrogenase in solid tumors and other diseases: Potential biomarkers and therapeutic targets. MedComm (Beijing) 2023; 4:e195. [PMID: 36694633 PMCID: PMC9842923 DOI: 10.1002/mco2.195] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
The family of aldehyde dehydrogenases (ALDHs) contains 19 isozymes and is involved in the oxidation of endogenous and exogenous aldehydes to carboxylic acids, which contributes to cellular and tissue homeostasis. ALDHs play essential parts in detoxification, biosynthesis, and antioxidants, which are of important value for cell proliferation, differentiation, and survival in normal body tissues. However, ALDHs are frequently dysregulated and associated with various diseases like Alzheimer's disease, Parkinson's disease, and especially solid tumors. Notably, the involvement of the ALDHs in tumor progression is responsible for the maintenance of the stem-cell-like phenotype, triggering rapid and aggressive clinical progressions. ALDHs have captured increasing attention as biomarkers for disease diagnosis and prognosis. Nevertheless, these require further longitudinal clinical studies in large populations for broad application. This review summarizes our current knowledge regarding ALDHs as potential biomarkers in tumors and several non-tumor diseases, as well as recent advances in our understanding of the functions and underlying molecular mechanisms of ALDHs in disease development. Finally, we discuss the therapeutic potential of ALDHs in diseases, especially in tumor therapy with an emphasis on their clinical implications.
Collapse
Affiliation(s)
- Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Siqin Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer MedicineNanjing Medical UniversityNanjingChina
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Darang E, Pezeshkian Z, Mirhoseini SZ, Ghovvati S. Identification of Key Genes and Potential Pathways Associated with Mastitis Induced by E. coli. Biochem Genet 2023; 61:202-220. [PMID: 35834114 PMCID: PMC9281188 DOI: 10.1007/s10528-022-10254-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/22/2022] [Indexed: 01/24/2023]
Abstract
Mastitis is one of the most important diseases of dairy cattle. It is an infectious disease leading to an inflammatory reaction in the cow's mammary gland. Escherichia coli is one of the common bacteria which induce mastitis in cows. The aim of this study was to identify key genes and potential pathways associated with mastitis induced by E. coli in dairy cattle using bioinformatics analysis. The gene expression profile of ten samples including five adjacent tissues from a quarter infected with Escherichia coli and five tissues from a healthy quarter of dairy cattle was assessed using GEO2R. Gene ontology and pathway analysis were performed using bioinformatics tools. A total of 156 differentially expressed genes were detected which 95 genes were upregulated and 61 genes were downregulated in adjacent tissue of quarter infected compared with healthy tissue. Cellular oxidant detoxification and oxidation-reduction process were the most significant biological process terms in gene ontology analysis. The most important pathways of DEGs were the biosynthesis of amino acids, p53 signaling pathway, and Metabolic pathways. Three important modules were identified and their path enrichment analysis was performed. There are 10 core genes, among which SOD2, COL1A2, COL3A1, POSTN, ALDH18A1, and CBS may be the main genes associated with mastitis, which can be considered as candidate genes in the prevention and carly diagnosis program of mastitis.
Collapse
Affiliation(s)
- Elham Darang
- Department of Animal Sciences, Faculty of Agriculture, University of Guilan, Rasht, 41635-1314, Guilan, Iran
| | - Zahra Pezeshkian
- Department of Animal Sciences, Faculty of Agriculture, University of Guilan, Rasht, 41635-1314, Guilan, Iran
| | - Seyed Ziaeddin Mirhoseini
- Department of Animal Sciences, Faculty of Agriculture, University of Guilan, Rasht, 41635-1314, Guilan, Iran
| | - Shahrokh Ghovvati
- Department of Animal Sciences, Faculty of Agriculture, University of Guilan, Rasht, 41635-1314, Guilan, Iran.
| |
Collapse
|
8
|
Yan T, Zhu M, Weng F, Zhu S, Wang C, Guo C. Comprehensive analysis of roles of atrial-fibrillation-related genes in lung adenocarcinoma using bioinformatic methods. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:55. [PMID: 36542177 DOI: 10.1007/s12032-022-01912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
Atrial fibrillation (AF) is the most common tachyarrhythmia in the world. Lung cancer is the leading cause of cancer deaths in 93 countries. Previous studies demonstrated that the prevalence of AF was higher in patients with lung cancer. However, research on the associations between AF and lung cancer is still rare. In the present study, we first identified AF-related genes using weighted gene correlation network analysis. We then analyzed the expression profiles, prognosis, immune infiltration, and methylation characteristics of these genes in LUAD patients using bioinformatics analysis. We found several AF-related genes, including CBX3, BUB1, DSC2, P4HA1, and CYP4Z1, which differently expressed between tumor and normal tissues. Survival analysis demonstrated that CYP4Z1 was positively correlated with overall survival in LUAD patients, while CBX3, BUB1, DSC2, and P4HA1 were negatively correlated. Moreover, we found that the methylation level of DSC2 in normal lung tissues was significantly higher than that in tumor tissues, and six methylation sites in the DNA sequences of DSC2 were identified negatively correlated with its expression levels. Immune infiltration analysis suggested that levels of immune cell infiltration were related to gene expression levels in varying degrees. We identified AF-related genes and found these genes were correlated with prognosis, immune infiltration, and methylation levels in lung cancer patients. We also constructed a risk signature based on these genes in LUAD patients. We hoped that the current study could provide a novel insight into roles of AF-related genes in lung cancer patients.
Collapse
Affiliation(s)
- Tao Yan
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Miao Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Fan Weng
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Shijie Zhu
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chunsheng Wang
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Changfa Guo
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
9
|
Kocher F, Puccini A, Untergasser G, Martowicz A, Zimmer K, Pircher A, Baca Y, Xiu J, Haybaeck J, Tymoszuk P, Goldberg RM, Petrillo A, Shields AF, Salem ME, Marshall JL, Hall M, Korn WM, Nabhan C, Battaglin F, Lenz HJ, Lou E, Choo SP, Toh CK, Gasteiger S, Pichler R, Wolf D, Seeber A. Multi-omic Characterization of Pancreatic Ductal Adenocarcinoma Relates CXCR4 mRNA Expression Levels to Potential Clinical Targets. Clin Cancer Res 2022; 28:4957-4967. [PMID: 36112544 PMCID: PMC9660543 DOI: 10.1158/1078-0432.ccr-22-0275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/13/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Chemokines are essential for immune cell trafficking and are considered to have a major impact on the composition of the tumor microenvironment. CX-chemokine receptor 4 (CXCR4) is associated with poor differentiation, metastasis, and prognosis in pancreatic ductal adenocarcinoma (PDAC). This study provides a comprehensive molecular portrait of PDAC according to CXCR4 mRNA expression levels. EXPERIMENTAL DESIGN The Cancer Genome Atlas database was used to explore molecular and immunologic features associated with CXCR4 mRNA expression in PDAC. A large real-word dataset (n = 3,647) served for validation and further exploratory analyses. Single-cell RNA analyses on a publicly available dataset and in-house multiplex immunofluorescence (mIF) experiments were performed to elaborate cellular localization of CXCR4. RESULTS High CXCR4 mRNA expression (CXCR4high) was associated with increased infiltration of regulatory T cells, CD8+ T cells, and macrophages, and upregulation of several immune-related genes, including immune checkpoint transcripts (e.g., TIGIT, CD274, PDCD1). Analysis of the validation cohort confirmed the CXCR4-dependent immunologic TME composition in PDAC irrespective of microsatellite instability-high/mismatch repair-deficient or tumor mutational burden. Single-cell RNA analysis and mIF revealed that CXCR4 was mainly expressed by macrophages and T-cell subsets. Clinical relevance of our finding is supported by an improved survival of CXCR4high PDAC. CONCLUSIONS High intratumoral CXCR4 mRNA expression is linked to a T cell- and macrophage-rich PDAC phenotype with high expression of inhibitory immune checkpoints. Thus, our findings might serve as a rationale to investigate CXCR4 as a predictive biomarker in patients with PDAC undergoing immune checkpoint inhibition.
Collapse
Affiliation(s)
- Florian Kocher
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - Alberto Puccini
- Medical Oncology Unit 1, Ospedale Policlinico San Martino, Genoa, Italy
| | - Gerold Untergasser
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - Agnieszka Martowicz
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - Kai Zimmer
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Pircher
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria.,Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Piotr Tymoszuk
- Data Analytics As a Service Tirol (DAAS) Tirol, Innsbruck, Austria
| | | | | | - Anthony F. Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Mohamed E. Salem
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina
| | - John L. Marshall
- Ruesch Center for The Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Michael Hall
- Department of Hematology and Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | | | | | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Emil Lou
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota
| | - Su-Pin Choo
- Curie Oncology, Mount Elizabeth Novena Specialist Centre, Singapore
| | - Chee-Keong Toh
- Curie Oncology, Mount Elizabeth Novena Specialist Centre, Singapore
| | - Silvia Gasteiger
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Seeber
- Department of Internal Medicine V (Hematology and Oncology), Comprehensive Cancer Center Innsbruck (CCCI), Medical University of Innsbruck, Innsbruck, Austria.,Corresponding Author: Andreas Seeber, Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria. Phone: 0043-50504-83166; E-mail:
| |
Collapse
|
10
|
Wang Z, Fan G, Zhu H, Yu L, She D, Wei Y, Huang J, Li T, Zhan S, Zhou S, Zhu Y, Wang Y, Chen X, Zhao J, Zhou G. PLOD2 high expression associates with immune infiltration and facilitates cancer progression in osteosarcoma. Front Oncol 2022; 12:980390. [PMID: 36276118 PMCID: PMC9581331 DOI: 10.3389/fonc.2022.980390] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/15/2022] [Indexed: 11/20/2022] Open
Abstract
Background Osteosarcoma (OS) is the most common primary malignant bone tumors in children and adolescents. Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) is a key gene in mediating the formation of the stabilized collagen cross-link, playing an important role in the progression of cancer. However, the interaction between OS and PLOD2 has not been clarified so far. Methods The target gene PLOD2 was screened through our own RNA-seq results and other two RNA-seq results from GEO database. The expression of PLOD2 in OS was detected by RT-qPCR, Western blot and immunohistochemistry. Functional experiments were performed to investigate the role of PLOD2 in OS cell invasion, migration and angiogenesis in vitro. An OS lung metastasis model was established to investigate the function of PLOD2 in OS metastasis and angiogenesis in vivo. The role of PLOD2 in immune infiltration in OS was explored by KEGG/GO analysis and immune infiltration analysis with TARGET, TCGA and TIMER. Results PLOD2 was high-expressed in OS, which was related to poor prognosis of OS patients. PLOD2 promoted OS cell migration, invasion and angiogenesis in vitro and aggravated OS metastasis and angiogenesis in vivo. Bioinformatic analysis showed that PLOD2 played an important role in immune cell infiltration in OS, including CD8 positive T cells, macrophages M0 cells, DC cells, endothelial cells, iDC cells, ly endothelial cells, MEP cells, mv endothelial cells, native B cells, smooth muscle cells and Th1 cells. Immunohistochemical results showed that the expression of CD4 and CD8A was negatively correlated with the expression of PLOD2 in OS. Conclusion PLOD2 was high-expressed in OS and promoted OS migration, invasion and angiogenesis in vitro and facilitated OS metastasis and angiogenesis in vivo. PLOD2 was associated with immune cell infiltration in OS, which could be a promising target to treat OS patients with metastasis and utilized to guide clinical immunotherapy in the future.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Gentao Fan
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Hao Zhu
- Department of Orthopaedics, Affiliated Jianhu Hospital of Nantong University, Yancheng, China
| | - Lingfeng Yu
- Department of Orthopaedics, Jinling Hospital, Nanjing University, Nanjing, China
| | - Diankun She
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Yanting Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jianhao Huang
- Department of Orthopaedics, Jinling Hospital, Southern Medical University, Nanjing, China
| | - Tianhang Li
- Department of Urology, Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Shoubin Zhan
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shenkai Zhou
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yan Zhu
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Yicun Wang
- Department of Orthopaedics, Jinling Hospital, Nanjing University, Nanjing, China
| | - Xi Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, China
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- *Correspondence: Guangxin Zhou, ; Jianning Zhao, ; Xi Chen,
| | - Jianning Zhao
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Guangxin Zhou, ; Jianning Zhao, ; Xi Chen,
| | - Guangxin Zhou
- Department of Orthopaedics, Jinling Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Guangxin Zhou, ; Jianning Zhao, ; Xi Chen,
| |
Collapse
|
11
|
Xu Q, Kong N, Zhao Y, Wu Q, Wang X, Xun X, Gao P. Pan-Cancer Analyses Reveal Oncogenic and Immunological Role of PLOD2. Front Genet 2022; 13:864655. [PMID: 35586565 PMCID: PMC9108334 DOI: 10.3389/fgene.2022.864655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022] Open
Abstract
Some previous studies have shown that PLOD2 has some value in tumorigenesis. However, the broad significance of PLOD2 has not been discussed in depth. This study was aimed at elaborated and summarized the value of PLOD2 in various tumors. First, we integrated GTEx, The Cancer Genome Atlas and Cancer Cell Line Encyclopedia databases to analyze the expression of PLOD2, and found that it was expressed differently in normal tissues and significantly highly expressed in most tumors compared with normal tissues. Second, our analysis revealed that PLOD2 expression was negatively correlated with the prognosis of several tumors. For gastric cancer, the median overall survival time was significantly higher in the PLOD2 low expression group [HR 0.616 (95%CI 0.442–0.858), p = 0.004]. Third, for tumor immunity, PLOD2 was significantly associated with tumor infiltration, including immune infiltrating cells; immune checkpoint expression; immune microenvironment scores (immune score, stromal score and estimate scores); immunotherapy-related scores (tumor mutational burden, microsatellite instability, tumor neoantigen burden); expression of DNA repair genes Mismatch Repairs and methyltransferase; and enrichment analyses identified PLOD2-associated terms and pathways. Lastly, twenty pairs of gastric cancer and adjacent immunohistochemistry showed that PLOD2 was significantly overexpressed in gastric cancer (p < 0.001). Collectively, PLOD2 played a significant role in tumorigenesis and maybe serve as a potential biomarker for diagnosis and prognosis in cancers.
Collapse
|
12
|
Abstract
Cancer is a complex disease and a significant cause of mortality worldwide. Over the course of nearly all cancer types, collagen within the tumor microenvironment influences emergence, progression, and metastasis. This review discusses collagen regulation within the tumor microenvironment, pathological involvement of collagen, and predictive values of collagen and related extracellular matrix components in main cancer types. A survey of predictive tests leveraging collagen assays using clinical cohorts is presented. A conclusion is that collagen has high predictive value in monitoring cancer processes and stratifying by outcomes. New approaches should be considered that continue to define molecular facets of collagen related to cancer.
Collapse
|
13
|
Benbrook DM. SHetA2 Attack on Mortalin and Colleagues in Cancer Therapy and Prevention. Front Cell Dev Biol 2022; 10:848682. [PMID: 35281109 PMCID: PMC8906462 DOI: 10.3389/fcell.2022.848682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Heat Shock Proteins of the 70-kDa family (HSP70s) do not cause cancer by themselves, but instead protect cells as they transform into cancer. These molecular chaperones bind numerous client proteins and utilize ATP hydrolysis to facilitate proper protein folding, formation of functional complexes and cellular localizations, or degradation of irreparably damaged proteins. Their transient upregulation by stressful situations avoids induction of programmed cell death. Continued upregulation of the mortalin, heat shock cognate (hsc70) and glucose regulated protein 78 (Grp78) support cancer development and progression by supporting pro-proliferative and metabolic functions and repressing pro-death functions of oncoproteins and tumor suppressor proteins. This review describes the discovery and development of a lead anti-cancer compound, sulfur heteroarotinoid A2 (SHetA2, NSC726189), which was originally developed to bind retinoic acid receptors, but was subsequently found to work independently of these receptors. The discovery and validation of mortalin, hsc70 and Grp78 as SHetA2 target proteins is summarized. The documented and hypothesized roles of these HSP70 proteins and their clients in the mechanism of SHetA2 inhibition of cancer without toxicity are discussed. Use of this mechanistic data to evaluate drug action in a cancer clinical trial and develop synergistic drug combinations is explained. Knowledge needed to optimize SHetA2 analogs for use in cancer therapy and prevention is proposed as future directions.
Collapse
|