1
|
Minoura H, Okamoto R, Hiki N, Yamashita K. Cancer-Associated Fibroblasts Genes and Transforming Growth Factor Beta Pathway in Gastric Cancer for Novel Therapeutic Strategy. Cancers (Basel) 2025; 17:795. [PMID: 40075643 PMCID: PMC11899367 DOI: 10.3390/cancers17050795] [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: 11/09/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 03/14/2025] Open
Abstract
Background-Objective: Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment of gastric cancer (GC). Understanding the molecular characteristics of CAFs-associated genes (CAFGs) is essential for elucidating their role in tumor progression and prognosis. This review aims to summarize the current knowledge on CAFGs, highlighting their expression patterns, prognostic significance, and potential functional mechanisms. Methods: A comprehensive review of existing literature was conducted, focusing on molecular features of CAFGs in GC. Single-cell RNA sequencing (scRNA-seq) analyses were examined to assess the expression patterns of CAFGs in broad-sense CAFs, which include both CAFs and pericytes. Additionally, clinicopathological studies validating the prognostic significance of CAFGs were reviewed. Results: ScRNA-seq analyses revealed that CAFGs are not necessarily restricted to CAFs alone but may also reflect the activation status of surrounding cells. Several CAFGs, including SPARC, THBS2, COL1A1, COL3A1, INHBA, PDGFC, and SDC2, have been validated for their prognostic relevance in GC. However, compared with other cancers, the functional mechanisms of these genes in GC remain poorly understood. While CAFGs exhibit synchronized expression with TGFB1 in colorectal cancer (CRC), such patterns have yet to be confirmed in GC due to the limitations of available microdissected data. Conclusions: A comprehensive understanding of CAFGs and their interaction with the TGFB pathway, including LTBP family genes, may be critical for developing novel therapeutic strategies for GC. Further research is needed to elucidate their functional mechanisms and therapeutic potential.
Collapse
Affiliation(s)
- Hiroyuki Minoura
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kitasato 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan; (H.M.); (R.O.)
| | - Riku Okamoto
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kitasato 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan; (H.M.); (R.O.)
| | - Naoki Hiki
- Department of Upper Gastrointestinal Surgery, Kitasato University School of Medicine, Kitasato 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan;
| | - Keishi Yamashita
- Division of Advanced Surgical Oncology, Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kitasato 1-15-1, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan; (H.M.); (R.O.)
| |
Collapse
|
2
|
Sun Y, Yu H, Han S, Ran R, Yang Y, Tang Y, Wang Y, Zhang W, Tang H, Fu B, Fu B, Weng X, Liu SM, Deng H, Peng S, Zhou X. Method for the extraction of circulating nucleic acids based on MOF reveals cell-free RNA signatures in liver cancer. Natl Sci Rev 2024; 11:nwae022. [PMID: 38348130 PMCID: PMC10860518 DOI: 10.1093/nsr/nwae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/24/2023] [Accepted: 09/24/2023] [Indexed: 02/15/2024] Open
Abstract
Cell-free RNA (cfRNA) allows assessment of health, status, and phenotype of a variety of human organs and is a potential biomarker to non-invasively diagnose numerous diseases. Nevertheless, there is a lack of highly efficient and bias-free cfRNA isolation technologies due to the low abundance and instability of cfRNA. Here, we developed a reproducible and high-efficiency isolation technology for different types of cell-free nucleic acids (containing cfRNA and viral RNA) in serum/plasma based on the inclusion of nucleic acids by metal-organic framework (MOF) materials, which greatly improved the isolation efficiency and was able to preserve RNA integrity compared with the most widely used research kit method. Importantly, the quality of cfRNA extracted by the MOF method is about 10-fold that of the kit method, and the MOF method isolates more than three times as many different RNA types as the kit method. The whole transcriptome mapping characteristics of cfRNA in serum from patients with liver cancer was described and a cfRNA signature with six cfRNAs was identified to diagnose liver cancer with high diagnostic efficiency (area under curve = 0.905 in the independent validation cohort) using this MOF method. Thus, this new MOF isolation technique will advance the field of liquid biopsy, with the potential to diagnose liver cancer.
Collapse
Affiliation(s)
- Yuqing Sun
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Haixin Yu
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shaoqing Han
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Ruoxi Ran
- Department of Clinical Laboratory, Center for Gene Diagnosis and Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ying Yang
- Department of Clinical Laboratory, Center for Gene Diagnosis and Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yongling Tang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Yuhao Wang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Wenhao Zhang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Heng Tang
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Boqiao Fu
- College of Chemistry and Materials Science, Hubei Engineering University, Xiaogan 432000, China
| | - Boshi Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xiaocheng Weng
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Song-Mei Liu
- Department of Clinical Laboratory, Center for Gene Diagnosis and Program of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hexiang Deng
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Shuang Peng
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Xiang Zhou
- College of Chemistry and Molecular Sciences, Key Laboratory of Biomedical Polymers-Ministry of Education, Wuhan University, Wuhan 430072, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
3
|
Jiang S, Sun HF, Li S, Zhang N, Chen JS, Liu JX. SPARC: a potential target for functional nanomaterials and drugs. Front Mol Biosci 2023; 10:1235428. [PMID: 37577749 PMCID: PMC10419254 DOI: 10.3389/fmolb.2023.1235428] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC), also termed osteonectin or BM-40, is a matricellular protein which regulates cell adhesion, extracellular matrix production, growth factor activity, and cell cycle. Although SPARC does not perform a structural function, it, however, modulates interactions between cells and the surrounding extracellular matrix due to its anti-proliferative and anti-adhesion properties. The overexpression of SPARC at sites, including injury, regeneration, obesity, cancer, and inflammation, reveals its application as a prospective target and therapeutic indicator in the treatment and assessment of disease. This article comprehensively summarizes the mechanism of SPARC overexpression in inflammation and tumors as well as the latest research progress of functional nanomaterials in the therapy of rheumatoid arthritis and tumors by manipulating SPARC as a new target. This article provides ideas for using functional nanomaterials to treat inflammatory diseases through the SPARC target. The purpose of this article is to provide a reference for ongoing disease research based on SPARC-targeted therapy.
Collapse
Affiliation(s)
- Shan Jiang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Hui-Feng Sun
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Shuang Li
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
- College Pharmacy, Jiamusi University, Jiamusi, China
| | - Ning Zhang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Ji-Song Chen
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
- School of Pharmaceutical Sciences, University of South China, Hengyang, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
4
|
Niu X, Ren L, Hu A, Zhang S, Qi H. Identification of Potential Diagnostic and Prognostic Biomarkers for Gastric Cancer Based on Bioinformatic Analysis. Front Genet 2022; 13:862105. [PMID: 35368700 PMCID: PMC8966486 DOI: 10.3389/fgene.2022.862105] [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: 01/25/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the most prevalent cancers all over the world. The molecular mechanisms of GC remain unclear and not well understood. GC cases are majorly diagnosed at the late stage, resulting in a poor prognosis. Advances in molecular biology techniques allow us to get a better understanding of precise molecular mechanisms and enable us to identify the key genes in the carcinogenesis and progression of GC. Methods: The present study used datasets from the GEO database to screen differentially expressed genes (DEGs) between GC and normal gastric tissues. GO and KEGG enrichments were utilized to analyze the function of DEGs. The STRING database and Cytoscape software were applied to generate protein–protein network and find hub genes. The expression levels of hub genes were evaluated using data from the TCGA database. Survival analysis was conducted to evaluate the prognostic value of hub genes. The GEPIA database was involved to correlate key gene expressions with the pathological stage. Also, ROC curves were constructed to assess the diagnostic value of key genes. Results: A total of 607 DEGs were identified using three GEO datasets. GO analysis showed that the DEGs were mainly enriched in extracellular structure and matrix organization, collagen fibril organization, extracellular matrix (ECM), and integrin binding. KEGG enrichment was mainly enriched in protein digestion and absorption, ECM-receptor interaction, and focal adhesion. Fifteen genes were identified as hub genes, one of which was excluded for no significant expression between tumor and normal tissues. COL1A1, COL5A2, P4HA3, and SPARC showed high values in prognosis and diagnosis of GC. Conclusion: We suggest COL1A1, COL5A2, P4HA3, and SPARC as biomarkers for the diagnosis and prognosis of GC.
Collapse
Affiliation(s)
- Xiaoji Niu
- Department of Gastroenterology of Traditional Chinese Medicine, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liman Ren
- Department of Endocrinology, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
| | - Aiyan Hu
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuhui Zhang
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Shuhui Zhang, ; Hongjun Qi,
| | - Hongjun Qi
- Department of Gastroenterology of Traditional Chinese Medicine, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
- *Correspondence: Shuhui Zhang, ; Hongjun Qi,
| |
Collapse
|
5
|
Wolf J, Hajdu RI, Boneva S, Schlecht A, Lapp T, Wacker K, Agostini H, Reinhard T, Auw-Hädrich C, Schlunck G, Lange C. Characterization of the Cellular Microenvironment and Novel Specific Biomarkers in Pterygia Using RNA Sequencing. Front Med (Lausanne) 2022; 8:714458. [PMID: 35174178 PMCID: PMC8841401 DOI: 10.3389/fmed.2021.714458] [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: 05/25/2021] [Accepted: 12/24/2021] [Indexed: 01/04/2023] Open
Abstract
With a worldwide prevalence of ~12%, pterygium is a common degenerative and environmentally triggered ocular surface disorder characterized by wing-shaped growth of conjunctival tissue onto the cornea that can lead to blindness if left untreated. This study characterizes the transcriptional profile and the cellular microenvironment of conjunctival pterygia and identifies novel pterygia-specific biomarkers. Formalin-fixed and paraffin-embedded pterygia as well as healthy conjunctival specimens were analyzed using MACE RNA sequencing (n = 8 each) and immunohistochemistry (pterygia n = 7, control n = 3). According to the bioinformatic cell type enrichment analysis using xCell, the cellular microenvironment of pterygia was characterized by an enrichment of myofibroblasts, T-lymphocytes and various antigen-presenting cells, including dendritic cells and macrophages. Differentially expressed genes that were increased in pterygia compared to control tissue were mainly involved in autophagy (including DCN, TMBIM6), cellular response to stress (including TPT1, DDX5) as well as fibroblast proliferation and epithelial to mesenchymal transition (including CTNNB1, TGFBR1, and FN1). Immunohistochemical analysis confirmed a significantly increased FN1 stromal immunoreactivity in pterygia when compared to control tissue. In addition, a variety of factors involved in apoptosis were significantly downregulated in pterygia, including LCN2, CTSD, and NISCH. Furthermore, 450 pterygia-specific biomarkers were identified by including transcriptional data of different ocular surface pathologies serving as controls (training group), which were then validated using transcriptional data of cultured human pterygium cells. Among the most pterygia-specific factors were transcripts such as AHNAK, RTN4, TPT1, FSTL1, and SPARC. Immunohistochemical validation of SPARC revealed a significantly increased stromal immunoreactivity in pterygia when compared to controls, most notably in vessels and intravascular vessel wall-adherent mononuclear cells. Taken together, the present study provides new insights into the cellular microenvironment and the transcriptional profile of pterygia, identifies new and specific biomarkers and in addition to fibrosis-related genes, uncovers autophagy, stress response and apoptosis modulation as pterygium-associated processes. These findings expand our understanding of the pathophysiology of pterygia, provide new diagnostic tools, and may enable new targeted therapeutic options for this common and sight-threatening ocular surface disease.
Collapse
Affiliation(s)
- Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Rozina Ida Hajdu
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Anja Schlecht
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Institute of Anatomy and Cell Biology, Wuerzburg University, Wuerzburg, Germany
| | - Thabo Lapp
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Katrin Wacker
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Thomas Reinhard
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Claudia Auw-Hädrich
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, Münster, Germany
- *Correspondence: Clemens Lange
| |
Collapse
|
6
|
Zhao X, Wu S, Jing J. Identifying Diagnostic and Prognostic Biomarkers and Candidate Therapeutic Drugs of Gastric Cancer Based on Transcriptomics and Single-Cell Sequencing. Pathol Oncol Res 2021; 27:1609955. [PMID: 34899080 PMCID: PMC8654733 DOI: 10.3389/pore.2021.1609955] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022]
Abstract
Background and Objective: Gastric cancer (GC) is an important health burden and the prognosis of GC is poor. We aimed to explore new diagnostic and prognostic indicators as well as potential therapeutic targets for GC in the current study. Methods: We screened the overlapped differentially expressed genes (DEGs) from GSE54129 and TCGA STAD datasets. Protein-protein interaction network analysis recognized the hub genes among the DEGs. The roles of these genes in diagnosis, prognosis, and their relationship with immune infiltrates and drug sensitivity of GC were analyzed using R studio. Finally, the clinically significant hub genes were verified using single-cell RNA sequencing (scRNA-seq) data. Results: A total of 222 overlapping genes were screened, which were enriched in extracellular matrix-related pathways. Further, 17 hub genes were identified, and our findings demonstrated that BGN, COMP, COL5A2, and SPARC might be important diagnostic and prognostic indicators of GC, which were also correlated with immune cell infiltration, tumor mutation burden (TMB), microsatellite instability (MSI), and sensitivity of therapeutic drugs. The scRNA-seq results further confirmed that all four hub genes were highly expressed in GC. Conclusion: Based on transcriptomics and single-cell sequencing, we identified four diagnostic and prognostic biomarkers of GC, including BGN, COMP, COL5A2, and SPARC, which can help predict drug sensitivity for GC as well.
Collapse
Affiliation(s)
- Xu Zhao
- Mathematical Computer Teaching and Research Office, Liaoning Vocational College of Medicine, Shenyang, China
| | - Shuang Wu
- College of Computer Science and Technology, Changchun Normal University, Changchun, China
| | - Jingjing Jing
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Proteomic Signatures of Diffuse and Intestinal Subtypes of Gastric Cancer. Cancers (Basel) 2021; 13:cancers13235930. [PMID: 34885041 PMCID: PMC8656738 DOI: 10.3390/cancers13235930] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a leading cause of death from cancer globally. Gastric cancer is classified into intestinal, diffuse and indeterminate subtypes based on histology according to the Laurén classification. The intestinal and diffuse subtypes, although different in histology, demographics and outcomes, are still treated in the same fashion. This study was designed to discover proteomic signatures of diffuse and intestinal subtypes. Mass spectrometry-based proteomics using tandem mass tags (TMT)-based multiplexed analysis was used to identify proteins in tumor tissues from patients with diffuse or intestinal gastric cancer with adjacent normal tissue control. A total of 7448 or 4846 proteins were identified from intestinal or diffuse subtype, respectively. This quantitative mass spectrometric analysis defined a proteomic signature of differential expression across the two subtypes, which included gremlin1 (GREM1), bcl-2-associated athanogene 2 (BAG2), olfactomedin 4 (OLFM4), thyroid hormone receptor interacting protein 6 (TRIP6) and melanoma-associated antigen 9 (MAGE-A9) proteins. Although GREM1, BAG2, OLFM4, TRIP6 and MAGE-A9 have all been previously implicated in tumor progression and metastasis, they have not been linked to intestinal or diffuse subtypes of gastric cancer. Using immunohistochemical labelling of a tissue microarray comprising of 124 cases of gastric cancer, we validated the proteomic signature obtained by mass spectrometry in the discovery cohort. Our findings should help investigate the pathogenesis of these gastric cancer subtypes and potentially lead to strategies for early diagnosis and treatment.
Collapse
|
8
|
Cao J, Gong J, Li X, Hu Z, Xu Y, Shi H, Li D, Liu G, Jie Y, Hu B, Chong Y. Unsupervised Hierarchical Clustering Identifies Immune Gene Subtypes in Gastric Cancer. Front Pharmacol 2021; 12:692454. [PMID: 34248641 PMCID: PMC8264374 DOI: 10.3389/fphar.2021.692454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023] Open
Abstract
Objectives: The pathogenesis of heterogeneity in gastric cancer (GC) is not clear and presents as a significant obstacle in providing effective drug treatment. We aimed to identify subtypes of GC and explore the underlying pathogenesis. Methods: We collected two microarray datasets from GEO (GSE84433 and GSE84426), performed an unsupervised cluster analysis based on gene expression patterns, and identified related immune and stromal cells. Then, we explored the possible molecular mechanisms of each subtype by functional enrichment analysis and identified related hub genes. Results: First, we identified three clusters of GC by unsupervised hierarchical clustering, with average silhouette width of 0.96, and also identified their related representative genes and immune cells. We validated our findings using dataset GSE84426. Subtypes associated with the highest mortality (subtype 2 in the training group and subtype C in the validation group) showed high expression of SPARC, COL3A1, and CCN. Both subtypes also showed high infiltration of fibroblasts, endothelial cells, hematopoietic stem cells, and a high stromal score. Furthermore, subtypes with the best prognosis (subtype 3 in the training group and subtype A in the validation group) showed high expression of FGL2, DLGAP1-AS5, and so on. Both subtypes also showed high infiltration of CD4+ T cells, CD8+ T cells, NK cells, pDC, macrophages, and CD4+ T effector memory cells. Conclusion: We found that GC can be classified into three subtypes based on gene expression patterns and cell composition. Findings of this study help us better understand the tumor microenvironment and immune milieu associated with heterogeneity in GC and provide practical information to guide personalized treatment.
Collapse
Affiliation(s)
- Jing Cao
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiao Gong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xinhua Li
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaoxia Hu
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yingjun Xu
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hong Shi
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Danyang Li
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guangjian Liu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yusheng Jie
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bo Hu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yutian Chong
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
9
|
Ran T, Chen Z, Zhao L, Ran W, Fan J, Hong S, Yang Z. LAMB1 Is Related to the T Stage and Indicates Poor Prognosis in Gastric Cancer. Technol Cancer Res Treat 2021; 20:15330338211004944. [PMID: 33784890 PMCID: PMC8020091 DOI: 10.1177/15330338211004944] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Gastric cancer (GC) is a common tumor malignancy with high incidence and poor prognosis. Laminin is an indispensable component of basement membrane and extracellular matrix, which is responsible for bridging the internal and external environment of cells and transmitting signals. This study mainly explored the association of the LAMB1 expression with clinicopathological characteristics and prognosis in gastric cancer. METHODS The expression data and clinical information of gastric cancer patients were downloaded from The Cancer Genome Atlas (TCGA) and Asian Cancer Research Group (ACRG). And we analyzed the relationship between LAMB1 expression and clinical characteristics through R. CIBERSORTx was used to calculate the absolute score of immune cells in gastric tumor tissues. Then COX proportional hazard models and Kaplan-Meier curves were performed to evaluate the role of LAMB1 and its influence on prognosis in gastric cancer patients. Finally, GO and KEGG analysis were applied for LAMB1-related genes in gastric cancer, and PPI network was constructed in Cytoscape software. RESULTS In the TCGA cohort, patients with gastric cancer frequently generated LAMB1 gene copy number variation, but had little effect on mRNA expression. Both in the TCGA and ACRG cohorts, the mRNA expression of LAMB1 in gastric cancer tissues was higher than it in normal tissues. All patients were divided into high expression group and low expression group according to the median expression level of LAMB1. The elevated expression group obviously had more advanced cases and higher infiltration levels of M2 macrophages. COX proportional hazard models and Kaplan-Meier curves revealed that patients with enhanced expression of LAMB1 have a worse prognosis. GO/KEGG analysis showed that LAMB1-related genes were enriched in PI3K-Akt signaling pathway, focal adhesion, ECM-receptor interaction, etc. CONCLUSIONS The high expression of LAMB1 in gastric cancer is related to the poor prognosis of patients, and it may be related to microenvironmental changes in tumors.
Collapse
Affiliation(s)
- Tao Ran
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - ZhiJi Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - LiWen Zhao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Wei Ran
- The Fourth Department of Infectious Disease, Chongqing Public Health Center, Chongqing, China
| | - JinYu Fan
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - SiYa Hong
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - ZhaoXia Yang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Shan Z, Wang W, Tong Y, Zhang J. Genome-Scale Analysis Identified NID2, SPARC, and MFAP2 as Prognosis Markers of Overall Survival in Gastric Cancer. Med Sci Monit 2021; 27:e929558. [PMID: 33758160 PMCID: PMC8006563 DOI: 10.12659/msm.929558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gastric cancer is the most common gastrointestinal tumor, and the rates of recurrence and metastasis are high. Research results on molecular biomarkers used for prognosis of gastric cancer remain inconclusive. This study aimed to explore the gene expression module of gastric cancer and to determine potential prognostic biomarkers. MATERIAL AND METHODS Three microarray datasets (GSE13911, GSE79973, and GSE29272) from Gene Expression Omnibus (GEO), including 206 pairs of gastric tumors and adjacent normal samples, were used for analysis of differentially expressed genes (DEGs). The 3 microarray datasets yielded 144 genes associated with the progression and prognosis of gastric cancer. After this, a risk score model was developed for result validation using an independent dataset from The Cancer Genome Atlas. RESULTS The validation of the independent dataset showed significantly increased NID2, SPARC, and MFAP2 expression in gastric tumor tissues, which were associated with poor outcomes in gastric cancer patients. Moreover, the high risk score obtained was associated with poor overall survival (HR: 1.787; 1.069-2.986; P=0.027). Subgroup analyses revealed that these significant prognostic values were detected in patients aged <65.0 years, tumors in the antrum/distal colon, grade 3 tumors, or TNM-M0 stages of cancer. CONCLUSIONS The findings of this study show that NID2, SPARC, and MFAP2 are upregulated in gastric tumor tissues and are significantly associated with poor overall survival. Therefore, the predictive values of the risk score model employed for the prognosis of gastric cancer could be improved by using these 3 upregulated DEGs.
Collapse
Affiliation(s)
- Zexing Shan
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Wentao Wang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Yilin Tong
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| | - Jianjun Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
11
|
Ritchie S, Reed DA, Pereira BA, Timpson P. The cancer cell secretome drives cooperative manipulation of the tumour microenvironment to accelerate tumourigenesis. Fac Rev 2021; 10:4. [PMID: 33659922 PMCID: PMC7894270 DOI: 10.12703/r/10-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cellular secretions are a fundamental aspect of cell-cell and cell-matrix interactions in vivo. In malignancy, cancer cells have an aberrant secretome compared to their non-malignant counterparts, termed the "cancer cell secretome". The cancer cell secretome can influence every stage of the tumourigenic cascade. At the primary site, cancer cells can secrete a multitude of factors that facilitate invasion into surrounding tissue, allowing interaction with the local tumour microenvironment (TME), driving tumour development and progression. In more advanced disease, the cancer cell secretome can be involved in extravasation and metastasis, including metastatic organotropism, pre-metastatic niche (PMN) preparation, and metastatic outgrowth. In this review, we will explore the latest advances in the field of cancer cell secretions, including its dynamic and complex role in activating the TME and potentiating invasion and metastasis, with comments on how these secretions may also promote therapy resistance.
Collapse
Affiliation(s)
- Shona Ritchie
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Daniel A Reed
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Brooke A Pereira
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
12
|
Bai J, Yang B, Shi R, Shao X, Yang Y, Wang F, Xiao J, Qu X, Liu Y, Zhang Y, Li Z. Could microtubule inhibitors be the best choice of therapy in gastric cancer with high immune activity: mutant DYNC1H1 as a biomarker. Aging (Albany NY) 2020; 12:25101-25119. [PMID: 33221769 PMCID: PMC7803585 DOI: 10.18632/aging.104084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
Abstract
Immune checkpoint blockade (ICB) has achieved unprecedented breakthroughs in various cancers, including gastric cancer (GC) with high immune activity (MSI-H or TMB-H), yet clinical benefits from ICB were moderate. Here we aimed to identify the most appropriate drugs which can improve outcomes in GC. We firstly compared MSI-H and TMB-H GC samples with normal samples in TCGA-STAD cohort, respectively. After that, Connectivity Map database repurposed nine candidate drugs (CMap score < -90). Then, microtubule inhibitors (MTIs) were screened as the significant candidate drugs with their representative gene sets strongly enriched (p < 0.05) via GSEA. GDSC database validated higher activities of some MTIs in GC cells with MSI-H and TMB-H (p < 0.05). Furthermore, some MTIs activities were positively associated with mutant Dynein Cytoplasmic 1 Heavy Chain 1 (DYNC1H1) (p < 0.05) based on NCI-60 cancer cell line panel. DYNC1H1 was high frequently alteration in GC and was positively associated with TMB-H and MSI-H. Mutant DYNC1H1 may be accompanied with down-regulation of MTIs-related genes in GC or change the binding pocket to sensitize MTIs. Overall, this study suggested that some MTIs may be the best candidate drugs to treat GC with high immune activity, especially patients with DYNC1H1 mutated.
Collapse
Affiliation(s)
- Jin Bai
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - BoWen Yang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Ruichuan Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Xinye Shao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Yujing Yang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Fang Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Jiawen Xiao
- Department of Medical Oncology, Shenyang Fifth People Hospital, Tiexi District, Shenyang 110001, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| | - Ye Zhang
- Laboratory I of Cancer Institute, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China.,Liaoning Province Clinical Research Center for Cancer, Shenyang 110001, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110001, China
| |
Collapse
|
13
|
Yin SP, Gao Y, Xie XS, Xu DD, Riabov V, Du WD. Accumulation of stabilin-1 positive macrophages in the early stage of gastric cancer is associated with short cumulative survival. Oncol Lett 2020; 19:2404-2412. [PMID: 32194740 PMCID: PMC7039161 DOI: 10.3892/ol.2020.11310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The scavenger receptor stabilin-1 has been reported to be expressed by tumor-associated macrophages (TAMs) and to facilitate tumor growth and metastasis in mouse models of breast carcinoma and melanoma. However, to the best of our knowledge, its expression and association with prognosis in human gastric cancer has not been evaluated. The present study investigated the expression of stabilin-1 and its association with clinicopathological parameters in patients with gastric cancer. The expression of stabilin-1 was evaluated by immunohistochemical staining of gastric cancer tissue samples of 371 Chinese patients with primary gastric adenocarcinoma. Confocal laser scanning microscopy was used to determine the cellular source of stabilin-1 in the gastric cancer tissues using anti-CD68, anti-CD163, anti-stabilin-1 and anti-secreted protein acidic and rich in cysteine antibodies. A higher number of stabilin-1-positive cells were observed in the cancer tissues of primary gastric adenocarcinoma compared with adjacent non-cancerous tissues of primary gastric adenocarcinoma (P<0.001); the majority of stabilin-1-positve cells were CD68+/CD163+ macrophages. Poorly-differentiated gastric cancer tissue had fewer stabilin-1-positive cells compared with medium- and well-differentiated gastric cancer (P=0.030). A higher number of stabilin-1-positive cells were found in the early Tumor-Node-Metastasis (TNM) stage (TNM I stage) of primary gastric adenocarcinoma (P=0.038) compared with TNM stage IV. For patients with TNM stage I disease, a higher number of stabilin-1-positive TAMs was associated with shorter cumulative survival (P<0.05). Overall, stabilin-1 was found to be expressed by CD68+ TAMs in human gastric cancer. The high expression of stabilin-1 in TNM stage I disease was associated with poor patient survival, indicating the clinical significance of stabilin-1 in gastric cancer.
Collapse
Affiliation(s)
- Shui-Ping Yin
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China.,Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yi Gao
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China.,Department of Biology, School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xu-Shi Xie
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Dan-Dan Xu
- Department of Oncology, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230012, P.R. China
| | - Vladimir Riabov
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, D-68167 Mannheim, Germany
| | - Wei-Dong Du
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China.,Department of Biology, School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
14
|
Xu B, Bai Z, Yin J, Zhang Z. Global transcriptomic analysis identifies SERPINE1 as a prognostic biomarker associated with epithelial-to-mesenchymal transition in gastric cancer. PeerJ 2019; 7:e7091. [PMID: 31218131 PMCID: PMC6563800 DOI: 10.7717/peerj.7091] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022] Open
Abstract
Background The plasminogen activation system plays a pivotal role in regulating tumorigenesis. In this work, we aim to identify key regulators of plasminogen activation associated with tumorigenesis and explore potential mechanisms in gastric cancer (GC). Methods Gene profiling datasets were extracted from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were screened for and obtained by the GEO2R tool. The Database for Annotation, Visualization and Integrated Discovery was used for GO and KEGG enrichment analysis. Gene set enrichment analysis (GSEA) was performed to verify molecular signatures and pathways among The Cancer Genome Atlas or GEO datasets. Correlations between SERPINE1 and markers of epithelial-to-mesenchymal transition (EMT) were analyzed using the GEPIA database and quantitative real-time PCR (qRT-PCR). Interactive networks of selected genes were built by STRING and Cytoscape software. Finally, selected genes were verified with the Kaplan–Meier (KM) plotter database. Results A total of 104 overlapped upregulated and 61 downregulated DEGs were obtained. Multiple GO and KEGG terms associated with the extracellular matrix were enriched among the DEGs. SERPINE1 was identified as the only regulator of angiogenesis and the plasminogen activator system among the DEGs. A high level of SERPINE1 was associated with a poor prognosis in GC. GSEA analysis showed a strong correlation between SERPINE1 and EMT, which was also confirmed with the GEPIA database and qRT-PCR validation. FN1, TIMP1, MMP2, and SPARC were correlated with SERPINE1.The KM plotter database showed that an overexpression of these genes correlated with a shorter survival time in GC patients. Conclusions In conclusion, SERPINE1 is a potent biomarker associated with EMT and a poor prognosis in GC. Furthermore, FN1, TIMP1, MMP2, and SPARC are correlated with SERPINE1 and may serve as therapeutic targets in reversing EMT in GC.
Collapse
Affiliation(s)
- Bodong Xu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Cancer Invasion and Metastasis Research, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Cancer Invasion and Metastasis Research, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jie Yin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Cancer Invasion and Metastasis Research, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Cancer Invasion and Metastasis Research, National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
15
|
Yu R, Li Z, Zhang C, Song H, Deng M, Sun L, Xu L, Che X, Hu X, Qu X, Liu Y, Zhang Y. Elevated limb-bud and heart development (LBH) expression indicates poor prognosis and promotes gastric cancer cell proliferation and invasion via upregulating Integrin/FAK/Akt pathway. PeerJ 2019; 7:e6885. [PMID: 31119084 PMCID: PMC6507893 DOI: 10.7717/peerj.6885] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 03/31/2019] [Indexed: 12/20/2022] Open
Abstract
The limb-bud and heart development (LBH) gene is a highly conserved, tissue-specific transcription cofactor in vertebrates that regulates multiple key genes in embryonic development. The role of LBH in various cancer types is still controversial, and its specific role and molecular mechanism in the oncogenesis of gastric cancer (GC) remains largely unexplored. In the present study, the prognostic significance and clinicopathological characteristics of LBH in GC was determined. The LBH mRNA expression was first investigated in four independent public datasets (TCGA-STAD, GSE15459, GSE29272, and GSE62254) and then validated with our samples at the protein level. LBH was overexpressed at both the mRNA and protein levels in cancer compared with normal tissues. High LBH expression was correlated with advanced T, N, and M stages. Kaplan–Meier analysis and log-rank test indicated that higher LBH expression was statistically correlated with shorter overall survival (OS) in the public datasets and our study samples. Univariate and multivariate Cox regression analysis showed that LBH was an independent prognostic biomarker for survival in TCGA-STAD, GSE15459, GSE62254 cohorts, and our GC patients. In vitro experiments showed that knockdown of LBH can significantly inhibit the proliferation and invasion of HGC-27 cells, while overexpression of LBH can significantly enhance the proliferation and invasion of BGC-823 cells. Gene Set Enrichment Analysis (GSEA), Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomics (KEGG) indicated that high LBH expression is associated with the PI3K-Akt pathway, focal adhesion, and extracellular matrix (ECM)-receptor interaction. Western blot analysis showed that knockdown of LBH significantly inhibited the expression of integrin α5, integrin β1, p-FAK, and p-Akt. Therefore, results from the present study indicate that LBH is a potential independent prognostic biomarker and promotes proliferation and invasion of GC cells by activating the integrin/FAK/Akt pathway.
Collapse
Affiliation(s)
- Ruoxi Yu
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Zhi Li
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Chuang Zhang
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Huicong Song
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Mingming Deng
- Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China.,Department of Respiratory and Infectious Disease of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ling Xu
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Xiaofang Che
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| | - Ye Zhang
- Department of Medical Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy, Shenyang, China
| |
Collapse
|
16
|
Lin X, Liu J, Hu SF, Hu X. Increased expression of TMED2 is an unfavorable prognostic factor in patients with breast cancer. Cancer Manag Res 2019; 11:2203-2214. [PMID: 31114314 PMCID: PMC6497492 DOI: 10.2147/cmar.s192949] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/19/2019] [Indexed: 12/24/2022] Open
Abstract
Background: We obtained 2 types of clones which were termed SC (sphere-shaped clone) and NSC (non-sphere-shaped clone) from 4T1 cells by monoclonal culture. SC and NSC were distinct in morphology, surface marker, metabolism and proliferation rate. With the transcriptome sequencing data analysis, we found TMED2 expressed higher in SCs. TMED2 was a member of the transmembrane emp24 domain and might play roles in cancer cell proliferation. However, its prognostic roles in breast cancer remained unknown. We aimed to investigate the prognostic values of TMED2 in patients with breast cancer. Methods: We used UALCAN (http://ualcan.path.uab.edu) and the Human Protein Atlas (www.proteinatlas.org) to explore the TMED2 expression level and DNA methylation data between breast cancer and normal breast tissue. With Oncomine (www.oncomine.org), we investigated the copy number of TMED2 in breast cancer sample and normal breast tissue. We used the Kaplan–Meier Plotter database (http://kmplot.com/analysis) to analyze prognostic values of TMED2 mRNA expression in all breast cancers and in different intrinsic subtypes. Moreover, protein expression levels of TMED2 were confirmed by Western blot in breast cancer tissues and normal mammary tissue as well as SCs and NSCs. Results: TMED2 significantly upregulated in breast cancer patients compared to normal mammary samples. Meanwhile, the increased expression of TMED2 mRNA was closely associated with reduced overall survival (OS) in all breast cancers, and with reduced OS in patients with ER-positive, Luminal A or Luminal B breast cancer subtypes. Moreover, western blot confirmed that TMED2 increased expressed was correlated with the reduced OS at protein levels. Conclusion: Increased expression of TMED2 was significantly related to unfavorable outcomes in patients with breast cancer. Thus, we supposed TMED2 is oncogenic and a potential target for breast cancer therapy and these preliminary findings require further study to determine whether TMED2-targeting reagents might be developed for clinical application in breast cancer.
Collapse
Affiliation(s)
- Xia Lin
- Cancer Institute (a Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jian Liu
- The Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Shu Fang Hu
- The Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Xun Hu
- Cancer Institute (a Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
17
|
Diagnostic role of circulating extracellular matrix-related proteins in non-small cell lung cancer. BMC Cancer 2018; 18:899. [PMID: 30227835 PMCID: PMC6145327 DOI: 10.1186/s12885-018-4772-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 08/23/2018] [Indexed: 12/16/2022] Open
Abstract
Background Interactions between cancer cells and the surrounding microenvironment are crucial determinants of cancer progression. During this process, bi-directional communication among tumor cells and cancer associated fibroblasts (CAF) regulate extracellular matrix (ECM) deposition and remodeling. As a result of this dynamic process, soluble ECM proteins can be released into the bloodstream and may represent novel circulating biomarkers useful for cancer diagnosis. The aim of the present study was to measure the levels of three circulating ECM related proteins (COL11A1, COL10A1 and SPARC) in plasma samples of lung cancer patients and in healthy heavy-smokers controls and test whether such measurements have diagnostic or prognostic value. Methods Gene expression profiling of lung fibroblasts isolated from paired normal and cancer tissue of NSCLC patients was performed by gene expression microarrays. The prioritization of the candidates for the study of circulating proteins in plasma was based on the most differentially expressed genes in cancer associated fibroblasts. Soluble ECM proteins were assessed by western blot in the conditioned medium of lung fibroblasts and by ELISA assays in plasma samples. Results Plasma samples from lung cancer patients and healthy heavy-smokers controls were tested for levels of COL11A1 and COL10A1 (n = 57 each) and SPARC (n = 90 each). Higher plasma levels of COL10A1 were detected in patients (p ≤ 0.001), a difference that was driven specifically by females (p < 0.001). No difference in COL11A1 levels between patients and controls was found. SPARC levels were also higher in plasma patients than controls (p < 0.001) with good performance in discriminating the two groups (AUC = 0.744). No significant association was observed between plasma proteins levels and clinicopathological features or survival. Conclusion Soluble factors related to proficient tumor-stroma cross-talk are detectable in plasma of primary lung cancer patients and may represent a valuable complementary diagnostic tool to discriminate lung cancer patients from healthy heavy-smokers individuals as shown for the SPARC protein. Electronic supplementary material The online version of this article (10.1186/s12885-018-4772-0) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Prognostic Value of Combination of Pretreatment Red Cell Distribution Width and Neutrophil-to-Lymphocyte Ratio in Patients with Gastric Cancer. Gastroenterol Res Pract 2018; 2018:8042838. [PMID: 29643918 PMCID: PMC5832175 DOI: 10.1155/2018/8042838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 12/20/2017] [Indexed: 12/14/2022] Open
Abstract
Aims Gastric cancer (GC) is often diagnosed at an advanced stage; inexpensive and valid biomarkers for GC are still unavailable. We aimed to evaluate the prognosis of the combination of pretreatment red cell distribution width (RDW) and neutrophil-to-lymphocyte ratio (NLR) in patients with GC. Methods A retrospective analysis from 103 GC patients who were diagnosed at our institution from 2012 to 2016 was performed. Both pretreatment RDW and NLR were calculated based on the recommended cutoff values of 13.4% and 2.755, respectively. Combined values of RDW and NLR (RDW + NLR) stratified patients into a score of 0 (RDW ≤ 13.4% and NLR ≤ 2.755), a score of 1 (RDW > 13.4% or NLR > 2.755), and a score of 2 (RDW > 13.4% and NLR > 2.755). Prognostic significances for overall survival (OS) and progression-free survival (PFS) were assessed. Results Pretreatment RDW + NLR was a significantly independent prognostic factor for OS and PFS. Moreover, high RDW + NLR was strongly related to age, tumor location, TNM stage, CA125, and CA199. In a subgroup analysis for patients with advanced gastric cancer (AGC), we observed that the level of RDW + NLR was markedly associated with OS and PFS. Conclusion Pretreatment RDW + NLR is a simple, inexpensive, and valid prognostic system to predict the survival in patients with GC, especially AGC.
Collapse
|
19
|
Peng P, Zhou X, Yi G, Chen P, Wang F, Dong W. Identification of a novel gene pairs signature in the prognosis of gastric cancer. Cancer Med 2018; 7:344-350. [PMID: 29282891 PMCID: PMC5806102 DOI: 10.1002/cam4.1303] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 11/25/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
Current prognostic signatures need to be improved in identifying high-risk patients of gastric cancer (GC). Thus, we aimed to develop a reliable prognostic signature that could assess the prognosis risk in GC patients. Two microarray datasets of GSE662254 (n = 300, training set) and GSE15459 (n = 192, test set) were included into analysis. Prognostic genes were screened to construct prognosis-related gene pairs (PRGPs). Then, a penalized Cox proportional hazards regression model identified seven PRGPs, which constructed a prognostic signature and divided patients into high- and low-risk groups according to the signature score. High-risk patients showed a poorer prognosis than low-risk patients in both the training set (hazard ratios [HR]: 6.086, 95% confidence interval [CI]: 4.341-8.533) and test set (1.773 [1.107-2.840]). The PRGPs signature also achieved a higher predictive accuracy (concordance index [C-index]: 0.872, 95% CI: 0.846-0.897) than two existing molecular signatures (0.706 [0.667-0.744] for a 11-gene signature and 0.684 [0.642-0.726] for a 24-lncRNA signature) and TNM stage (0.764 [0.715-0.814]). In conclusion, our study identified a novel gene pairs signature in the prognosis of GC.
Collapse
Affiliation(s)
- Pai‐Lan Peng
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhan430060China
- Department of GastroenterologyThe Central Hospital of Enshi Autonomous PrefectureEnshi445000China
| | - Xiang‐Yu Zhou
- Department of GastroenterologyThe Central Hospital of Enshi Autonomous PrefectureEnshi445000China
| | - Guo‐Dong Yi
- Department of GastroenterologyThe Central Hospital of Enshi Autonomous PrefectureEnshi445000China
| | - Peng‐Fei Chen
- Department of GastroenterologyThe Central Hospital of Enshi Autonomous PrefectureEnshi445000China
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Fan Wang
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Wei‐Guo Dong
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhan430060China
| |
Collapse
|
20
|
Zarogoulidis P, Huang H, Bai C, Petridis D, Papadopoulou S, Faniadou E, Eleftheriadou E, Trakada G, Cristoforos K, Rapti A, Yarmus L, Kopman DF, Man YG, Hohenforst-Schmidt W. Nab-paclitaxel as First Line Treatment for NSCLC in Elderly Patients More Than 75 Years Old. J Cancer 2017; 8:1673-1678. [PMID: 28775787 PMCID: PMC5535723 DOI: 10.7150/jca.19463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 05/03/2017] [Indexed: 12/30/2022] Open
Abstract
Introduction: Lung cancer is still the leading cause of cancer among cancer patients. Although there are novel therapies as second line treatment for NSCLC, there is an issue for elderly patients. Patients and Methods: We collected retrospectively data from 60 patients >75 years of age. Thirty of these patients received nab-paclitaxel and first line treatment and were compared to thirty patients that received only best supportive care. Results: The median life of patients at the date of disease progression, although increased by the administration of the drug (92 days versus 70) was not confirmed statistically significantly (Mann-Whitney test: W = 280, p = 0.138). The administration of drug seems to keep stable the biological condition of patients (McNemar's test: χ2 = 0.033, p = 0.99). Patients with chemotherapy the death rate was increased by 50% as compared to those with best supportive care (12 vs 8), the median life until the unfortunate event surpassed statistically significantly the latter (150 days of life as compared to 108, Mann-Whitney test: W = 57.5, p = 0.045). Discussion: Nab-paclitaxel as a monotherapy could be considered as a first line treatment option for patients > 75 years of age without any previous cardiological medical history when compared to best supportive care.
Collapse
Affiliation(s)
- Paul Zarogoulidis
- Pulmonary Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Haidong Huang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Dimitris Petridis
- Department of Food Technology, School of Food Technology and Nutrition, Alexander Technological Educational Institute, Thessaloniki, Greece
| | - Susana Papadopoulou
- Department of Nutrition and Dietetics, Alexander Technological Educational Institute, Thessaloniki, Greece
| | - Eleni Faniadou
- Pulmonary Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ellada Eleftheriadou
- Pulmonary Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Trakada
- Division of Pulmonology, Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Alexandra Hospital, Athens, Greece
| | - Kosmidis Cristoforos
- General Surgery Department, European Interbalkan Medical Center, Thessaloniki, Greece
| | - Aggeliki Rapti
- Second Pulmonary Clinic, 'Sotiria' Chest Diseases Hospital, Athens, Greece
| | - Lonny Yarmus
- Division of Pulmonary and Critical Care Medicine, Sheikh Zayed Cardiovascular & Critical Care Tower, Baltimore, U.S.A
| | - David-Feller Kopman
- Division of Pulmonary and Critical Care Medicine, Sheikh Zayed Cardiovascular & Critical Care Tower, Baltimore, U.S.A
| | - Yan-Gao Man
- Research Laboratory and International Collaboration, Bon Secours Cancer Institute, VA, USA
| | | |
Collapse
|