1
|
Piyarathne N, Hettiarachchi K, Yuwanati M, Sivaramakrishnan G, Ramanathan A, Jayasinghe R, Chala S, Chaurasia A. Osteopontin as a prognostic biomarker in head and neck cancer- a systematic review and meta-analysis. Evid Based Dent 2025:10.1038/s41432-025-01133-8. [PMID: 40074805 DOI: 10.1038/s41432-025-01133-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/30/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Osteopontin (OPN) is linked to cancer progression, metastasis, and treatment resistance in head and neck cancer (HNC). This meta-analysis evaluated OPN as a prognostic biomarker in HNC. METHOD A comprehensive search was conducted for studies published up to December 2023, including English papers on HNC analyzing OPN expression. Data extraction, quality assessment, and quantitative analysis were performed using fixed and random effect models with 95% CI. Heterogeneity and publication bias were assessed with I2 and Egger's regression test. RESULTS Fifty-one studies were included. OPN expression was significantly elevated in tissue and plasma in HNC compared to control (SMD 0.98; 95% CI 0.47-1.49; I2 = 13%; p < 0.00). High plasma OPN predicted poor survival (HR: 2.00; I2 = 64%; P = 0.03), as did high tissue OPN (hazard ratio: 2.71, 95% confidence interval = 1.51-4.87; I2 = 49.4%, p > 0.05). Elevated plasma OPN correlated with smoking, poorly differentiated neoplasms, larger tumors, advanced stage, and lymph node metastasis. Positive tissue OPN was associated with nodal involvement, advanced stage, male gender, and smoking. CONCLUSION OPN is a robust prognostic biomarker in HNC, indicating tumor aggressiveness and poor prognostic outcomes. Standardized measurement protocols and further validation in prospective studies are necessary.
Collapse
Affiliation(s)
- Nadisha Piyarathne
- Department of Basic Sciences, Center for Research in Oral Cancer (CROC), Faculty of Dental Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| | - Kalani Hettiarachchi
- Frazer Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Monal Yuwanati
- Department of Oral and Maxillofacial pathology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | | | - Anand Ramanathan
- Department of Oral & Maxillofacial Clinical Sciences, Faculty of Dentistry, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ruwan Jayasinghe
- Department of Oral Medicine and Periodontology, Faculty of Dental Sciences, University of Peradeniya, Peradeniya, Sri Lanka
| | - Sanaa Chala
- Faculty of Dental Medicine, Mohammed V University in Rabat, Rue Mohammed Jazouli, BP, 6212 Madinat Al Irfane, Rabat, Morocco
- Laboratory of Biostatistics, Clinical and Epidemiological Research, Faculty of Medicine and Pharmacy, University Mohammed V in Rabat, Rabat, Morocco
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, King George's Medical University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
2
|
Mattos D, Rocha M, Tessmann J, Ferreira L, Gimba E. Overexpression of Osteopontin-a and Osteopontin-c Splice Variants Are Worse Prognostic Features in Colorectal Cancer. Diagnostics (Basel) 2024; 14:2108. [PMID: 39410512 PMCID: PMC11475046 DOI: 10.3390/diagnostics14192108] [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: 08/06/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Osteopontin (OPN) is a glycoprotein involved in various physiological and pathological processes, and its aberrant expression in cancer cells is closely linked to tumor progression. In colorectal cancer (CRC), OPN is overexpressed, but the roles of its splice variants (OPN-SVs), OPNa, OPNb, and OPNc, are not well understood. This study aimed to characterize the expression patterns of OPN-SVs and their potential diagnostic and prognostic implications in CRC using transcriptomic data deposited in TSVdb and TCGA. Methods: The expression patterns of each OPN-SV were analyzed using transcriptomic data deposited in TSVdb and TCGA, which were correlated to patient data available at cBioPortal. Results: Bioinformatic analysis revealed that OPNa, OPNb, and OPNc are overexpressed in CRC samples compared to non-tumor samples. Notably, OPNa and OPNc are overexpressed in CRC stages (II, III, and IV) compared to stage I. Higher levels of OPNa and OPNc transcripts are associated with worse overall survival (OS) and shorter progression-free survival (PFS) in CRC patients. Additionally, the expression of OPNa, OPNb, and OPNc is correlated with BRAFV600E mutations in CRC samples. Conclusions: These findings suggest that OPNa and OPNc, in particular, have potential as diagnostic and prognostic biomarkers, paving the way for their further evaluation in CRC diagnosis and prognosis.
Collapse
Affiliation(s)
- Daniella Mattos
- Hemato-Oncology Molecular Program, National Institute of Cancer, 23rd Red Cross Square, 6th Floor, Rio de Janeiro 20230-130, RJ, Brazil;
- Biomedical Science Graduation Program, Fluminense Federal University, Rua Professor Hernani Pires de Melo, 101, Niterói 24210-130, RJ, Brazil
| | - Murilo Rocha
- Cellular and Molecular Oncobiology Program, National Institute of Cancer, Rio de Janeiro 20231-050, RJ, Brazil; (M.R.); (J.T.)
| | - Josiane Tessmann
- Cellular and Molecular Oncobiology Program, National Institute of Cancer, Rio de Janeiro 20231-050, RJ, Brazil; (M.R.); (J.T.)
| | - Luciana Ferreira
- Hemato-Oncology Molecular Program, National Institute of Cancer, 23rd Red Cross Square, 6th Floor, Rio de Janeiro 20230-130, RJ, Brazil;
- Departamento de Genética, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro, BR-465, Km 07, Seropédica, Rio de Janeiro 23897-000, RJ, Brazil
| | - Etel Gimba
- Hemato-Oncology Molecular Program, National Institute of Cancer, 23rd Red Cross Square, 6th Floor, Rio de Janeiro 20230-130, RJ, Brazil;
- Biomedical Science Graduation Program, Fluminense Federal University, Rua Professor Hernani Pires de Melo, 101, Niterói 24210-130, RJ, Brazil
- Departamento de Ciências da Natureza, Humanities and Healthy Institute, Fluminense Federal University, Recife Street, Bela Vista, Rio das Ostras 28895-532, RJ, Brazil
| |
Collapse
|
3
|
Jin C, Liao S, Lu G, Geng BD, Ye Z, Xu J, Ge G, Yang D. Cellular senescence in metastatic prostate cancer: A therapeutic opportunity or challenge (Review). Mol Med Rep 2024; 30:162. [PMID: 38994760 PMCID: PMC11258599 DOI: 10.3892/mmr.2024.13286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/14/2024] [Indexed: 07/13/2024] Open
Abstract
The treatment of patients with metastatic prostate cancer (PCa) is considered to be a long‑standing challenge. Conventional treatments for metastatic PCa, such as radical prostatectomy, radiotherapy and androgen receptor‑targeted therapy, induce senescence of PCa cells to a certain extent. While senescent cells can impede tumor growth through the restriction of cell proliferation and increasing immune clearance, the senescent microenvironment may concurrently stimulate the secretion of a senescence‑associated secretory phenotype and diminish immune cell function, which promotes PCa recurrence and metastasis. Resistance to established therapies is the primary obstacle in treating metastatic PCa as it can lead to progression towards an incurable state of disease. Therefore, understanding the molecular mechanisms that underly the progression of PCa is crucial for the development of novel therapeutic approaches. The present study reviews the phenomenon of treatment‑induced senescence in PCa, the dual role of senescence in PCa treatments and the mechanisms through which senescence promotes PCa metastasis. Furthermore, the present review discusses potential therapeutic strategies to target the aforementioned processes with the aim of providing insights into the evolving therapeutic landscape for the treatment of metastatic PCa.
Collapse
Affiliation(s)
- Cen Jin
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
- Medical Imaging School, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Sijian Liao
- Clinical Medicine School, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Guoliang Lu
- Department of Pediatrics, Anshun People's Hospital, Anshun, Guizhou 561000, P.R. China
| | - Bill D. Geng
- School of Natural Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Zi Ye
- Clinical Medicine School, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Jianwei Xu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Guo Ge
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| | - Dan Yang
- Department of Surgery, Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou 561113, P.R. China
| |
Collapse
|
4
|
Chai Z, Zhu C, Wang X, Zheng Y, Han F, Xie Q, Liu C. PADI3 inhibits epithelial-mesenchymal transition by targeting CKS1-induced signal transduction in colon cancer. J Cancer Res Ther 2024; 20:1323-1333. [PMID: 39206995 DOI: 10.4103/jcrt.jcrt_558_24] [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: 03/21/2024] [Accepted: 06/03/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Protein arginine deiminase 3 (PADI3) is involved in various biological processes of human disease. PADI3 has recently received increasing attention due to its role in tumorigenesis. In a previous study, we found that PADI3 plays a tumor suppressor role in colon cancer by inducing cell cycle arrest, but its critical role and mechanism in cancer metastasis remain obscure. In this study, we fully studied the role of PADI3 in colon cancer cell metastasis. METHODS The expression levels of related proteins were detected by Western blotting, and Transwell and wound healing assays were used to examine the cell migration ability. Flow cytometry was used to measure and exclude cell apoptosis-affected cell migration. Both overexpression and rescue experiments were employed to elucidate the molecular mechanism of CKS1 in colon cancer cells. RESULTS The expression levels of PADI3 and CKS1 are negatively related, and PADI3 can promote CKS1 degradation in a ubiquitin-dependent manner. PADI3 can suppress colon cancer cell migration and reduce the wound healing speed by inhibiting CKS1 expression. The molecular mechanism showed that CKS1 can promote EMT by increasing Snail and N-cadherin expression and suppressing E-cadherin expression. PADI3, as a suppressor of CKS1, can block the process of EMT by impairing CKS1-induced Snail upregulation and E-cadherin downregulation; however, the expression of N-cadherin cannot be rescued. CONCLUSIONS CKS1 promotes EMT in colon cancer by regulating Snail/E-cadherin expression, and this effect can be reversed by PADI3 via the promotion of CKS1 degradation in a ubiquitylation-dependent manner.
Collapse
Affiliation(s)
- Zhengbin Chai
- Translational Research Laboratory for Stem Cell and Traditional Chinese Medicine, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
- Department of Clinical Laboratory Medicine, Shandong Public Health Clinical Center, Shandong University, Jinan, People's Republic of China
| | - Changhui Zhu
- Department of Biochemistry and Molecular Biology, Shandong Provincial Qianfoshan Hospital, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, People's Republic of China
| | - Xiwei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Yingying Zheng
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Fabin Han
- Translational Research Laboratory for Stem Cell and Traditional Chinese Medicine, Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People's Republic of China
| | - Qi Xie
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Chunyan Liu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People's Republic of China
- Department of Plastic Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, People's Republic of China
| |
Collapse
|
5
|
Zheng S, He H, Zheng J, Zhu X, Lin N, Wu Q, Wei E, Weng C, Chen S, Huang X, Jian C, Guan S, Yang C. Machine learning-based screening and validation of liver metastasis-specific genes in colorectal cancer. Sci Rep 2024; 14:17679. [PMID: 39085446 PMCID: PMC11291988 DOI: 10.1038/s41598-024-68706-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Colorectal liver metastasis (CRLM) is challenging in the clinical treatment of colorectal cancer. Limited research has been conducted on how CRLM develops. RNA sequencing data were obtained from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). Four machine learning algorithms were used to screen the hub CRLM-specific genes, including Least Absolute Shrinkage and Selection Operator (Lasso), Random forest, SVM-RFE, and XGboost. The model for identifying CRLM was developed using stepwise logistic regression and was validated using internal and independent datasets. The prognostic value of hub CRLM-specific genes was assessed using the Lasso-Cox method. The in vitro experiments were performed using SW620 cells. The CRLM identification model was developed based on four CRLM-specific genes (SPP1, ZG16, P2RY14, and PRKAR2B), and the model efficacy was validated using GSE41258 and three external cohorts. Five CRLM-specific prognostic hub genes, SPP1, ZG16, P2RY14, CYP2E1, and C5, were identified using the Lasso-Cox algorithm, and a risk score was constructed. The risk score was validated using the GSE39582 cohort. Three genes have both efficacy in identifying CRLM and prognostic value: ZG16, P2RY14, and SPP1. Immune infiltration and enrichment analyses demonstrated that SPP1 was associated with M2 macrophage polarization and extracellular matrix remodeling. In vitro experiments indicated that SPP1 may act as a cancer-promoting factor. The hub CRLM-specific gene SPP1 can help determine the diagnosis, prognosis, and immune infiltration of patients with CRLM.
Collapse
Affiliation(s)
- Shiyao Zheng
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Hongxin He
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Jianfeng Zheng
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Xingshu Zhu
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, 350025, People's Republic of China
| | - Nan Lin
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, 350025, People's Republic of China
- Fuzong Clinical Medical College of Fujian Medical University, Department of General Surgery, 900th Hospital of Joint Logistics Support Force, PLA, Fuzhou, 350025, People's Republic of China
| | - Qing Wu
- Department of Oncology, Molecular Oncology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, People's Republic of China
| | - Enhao Wei
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Caiming Weng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350002, People's Republic of China
| | - Shuqian Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, People's Republic of China
| | - Xinxiang Huang
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Chenxing Jian
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350108, People's Republic of China.
- Department of Anorectal Surgery, Afliated Hospital of Putian University, Putian, 351106, People's Republic of China.
| | - Shen Guan
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China.
| | - Chunkang Yang
- Department of Gastrointestinal Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China.
- Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, People's Republic of China.
| |
Collapse
|
6
|
Fukusada S, Shimura T, Natsume M, Nishigaki R, Okuda Y, Iwasaki H, Sugimura N, Kitagawa M, Katano T, Tanaka M, Ozeki K, Kubota E, Hayashi K, Kataoka H. Osteopontin secreted from obese adipocytes enhances angiogenesis and promotes progression of pancreatic ductal adenocarcinoma in obesity. Cell Oncol (Dordr) 2024; 47:229-244. [PMID: 37640984 DOI: 10.1007/s13402-023-00865-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2023] [Indexed: 08/31/2023] Open
Abstract
PURPOSE Obesity is a risk factor and poor prognostic factor for pancreatic ductal adenocarcinoma (PDAC), but the underlying mechanisms remain unclear. METHODS PDAC cells and obese visceral adipocytes (O-Ad) derived from mice and humans were used to analyze interactions between the two cell types, and human microvascular endothelial cells were used for angiogenesis assay. A xenograft mouse model with subcutaneously injected PDAC cells was used for animal studies. The relationship between visceral fat and prognosis was analyzed using resected tissues from PDAC patients with and without obesity. RESULTS Conditioned media (CM) from O-Ad significantly increased PDAC cell growth and migration and angiogenic capacity in both human and mice cells, and blocking osteopontin (OPN) in O-Ad canceled O-Ad-induced effects in both mouse and human cells. In addition, O-Ad directly increased the migratory and tube-forming capacities of endothelial cells, while blocking OPN canceled these effects. O-Ad increased AKT phosphorylation and VEGFA expression in both PDAC and endothelial cells, and OPN inhibition in O-Ad canceled those O-Ad-induced effects. In the xenograft model, PDAC tumor volume was significantly increased in obese mice compared with lean mice, whereas blocking OPN significantly inhibited obesity-accelerated tumor growth. OPN expression in adipose tissues adjacent to human PDAC tumor was significantly higher in obese patients than in non-obese patients. In PDAC patients with obesity, high OPN expression in adipose tissues was significantly associated with poor prognosis. CONCLUSION Obese adipocytes trigger aggressive transformation in PDAC cells to induce PDAC progression and accelerate angiogenesis via OPN secretion.
Collapse
Affiliation(s)
- Shigeki Fukusada
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Makoto Natsume
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Ruriko Nishigaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Yusuke Okuda
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiroyasu Iwasaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Naomi Sugimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Mika Kitagawa
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takahito Katano
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Mamoru Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Keiji Ozeki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Eiji Kubota
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Kazuki Hayashi
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| |
Collapse
|
7
|
Zhu X, Ji J, Han X. Osteopontin: an essential regulatory protein in idiopathic pulmonary fibrosis. J Mol Histol 2024; 55:1-13. [PMID: 37878112 DOI: 10.1007/s10735-023-10169-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, chronic lung disease characterized by abnormal proliferation and activation of fibroblasts, excessive accumulation of extracellular matrix (ECM), inflammatory damage, and disrupted alveolar structure. Despite its increasing morbidity and mortality rates, effective clinical treatments for IPF remain elusive. Osteopontin (OPN), a multifunctional ECM protein found in various tissues, has been implicated in numerous biological processes such as bone remodeling, innate immunity, acute and chronic inflammation, and cancer. Recent studies have highlighted the pivotal role of OPN in the pathogenesis of IPF. This review aims to delve into the involvement of OPN in the inflammatory response, ECM deposition, and epithelial-mesenchymal transition (EMT) during IPF, and intends to lay a solid theoretical groundwork for the development of therapeutic strategies for IPF.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Jie Ji
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
8
|
Jiang N, Jin L, Li S. Role of SPP1 in the diagnosis of gastrointestinal cancer. Oncol Lett 2023; 26:411. [PMID: 37614657 PMCID: PMC10442757 DOI: 10.3892/ol.2023.13997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/06/2023] [Indexed: 08/25/2023] Open
Abstract
Recently, the incidence rate of digestive system tumors has increased in China and these tumors occur in a younger population. The present study aimed to determine the expression levels and potential clinical value of secreted phosphoprotein 1 (SPP1) in gastrointestinal cancer. The microarray datasets GSE104836, GSE189830 and GSE103236, obtained from the gene expression omnibus database, were analyzed to determine differentially expressed genes in patients with colorectal cancer (CRC), gastric cancer (GC) and esophageal cancer (EC). A total of 42 patients with CRC, GC or EC and 21 healthy controls were recruited to obtain blood and tissues samples. SPP1 expression levels were detected using reverse transcription-quantitative PCR. Moreover, levels of significance of SPP1 in patients with CRC, GC and EC were analyzed using receiver operating characteristic analysis. Potential correlations between SPP1 and carcinoembryonic antigen (CEA) were assessed using Pearson's correlation coefficient. SPP1 was significantly upregulated in the serum, plasma and tissue of patients with CRC, GC or EC. In addition, the area under the curve of SPP1 was >0.5 in the plasma, serum and cancer tissue of patients with early and late CRC, GC or EC. The present study further demonstrated that the specificity and sensitivity of SPP1 was higher in patients with late CRC, GC or EC compared with patients with early CRC, GC or EC. Moreover, SPP1 and CEA were significantly positively correlated in serum of patients with CRC, GC or EC. In conclusion, the current study demonstrated that SPP1 exhibited significant diagnostic value for gastrointestinal tumors, which suggested that SPP1 may exhibit potential as a diagnostic marker of CRC, GC and EC. The present study provided a novel theoretical basis for the role of SPP1 as a diagnostic marker of digestive system tumors.
Collapse
Affiliation(s)
- Nanfang Jiang
- Department of Gastroenterology, Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei 430030, P.R. China
| | - Lei Jin
- Department of Gastroenterology, Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei 430030, P.R. China
| | - Shuyu Li
- Department of Gastroenterology, Hubei No. 3 People's Hospital of Jianghan University, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
9
|
OPN Promotes Cell Proliferation and Invasion through NF- κB in Human Esophageal Squamous Cell Carcinoma. Genet Res (Camb) 2022; 2022:3154827. [PMID: 36619897 PMCID: PMC9779994 DOI: 10.1155/2022/3154827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
Background Osteopontin (OPN) is a phosphorylated glycoprotein. There is increasing evidence that the OPN gene played a major role in the progression of solid organ tumors. However, few studies have clarified how OPN regulated the functional role of human esophageal squamous cell carcinoma (ESCC). This study was designed to investigate the effect of OPN in esophageal squamous cell carcinoma. Methods First, we screened Eca-109 and KYSE-510 cells to construct OPN silencing and overexpression models. Endogenous OPN of Eca-109 and KYSE-510 were knocked down or overexpressed using small interfering RNAs. QRT-PCR, Western blot, flow cytometry, and CCK-8 were used to detect the function of Eca-109 and KYSE-510 cells. Tumor formation in nude mice was used to measure tumor growth after OPN inhibition. Results Eca-109 and KYSE-510 cells contain the si-OPN arrest cell cycle in the S-phase and increase apoptosis. These changes were OPN downregulation of the NF-κB pathway that significantly reduced the protein levels of TNF-α, IL-1β, and p-p65. However, the activity of Eca-109 and KYSE-510 cells was enhanced in OPN overexpressing cells. Then, the in vivo tumor formation experiment in nude mice showed that the tumor volume and weight of nude mice after silencing OPN were significantly reduced. Conclusion This study contributed to understanding the vital role of OPN in ESCC development and progression. This could be a promising molecular target for developing new ESCC diagnostic and therapeutic strategies.
Collapse
|
10
|
Xu J, Yang M, Shao AZ, Pan HW, Fan YX, Chen KP. Identification and Validation of Common Reference Genes for Normalization of Esophageal Squamous Cell Carcinoma Gene Expression Profiles. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9125242. [PMID: 36467891 PMCID: PMC9711964 DOI: 10.1155/2022/9125242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 09/04/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the subtypes of esophageal cancer with Chinese characteristics, and its five-year survival rate is less than 20%. Early diagnosis is beneficial to improving the survival rate of ESCC significantly. Quantitative Real-Time Polymerase Chain Reaction is a high-throughput technique that can quantify tumor-related genes for early diagnosis. Its accuracy largely depends on the stability of the reference gene. There is no systematic scientific basis to demonstrate which reference gene expression is stable in ESCC and no consensus on the selection of internal reference. Therefore, this research used four software programs (The comparative delta-Ct method, GeNorm, NormFinder, and BestKeeper) to evaluate the expression stability of eight candidate reference genes commonly used in other tumor tissues and generated a comprehensive analysis by RefFinder. Randomly selected transcriptome sequencing analysis confirmed the SPP1 gene is closely related to ESCC. It was found that the expression trend of SPP1 obtained by RPS18 and PPIA as internal reference genes were the same as that of sequencing. The results show that RPS18 and PPIA are stable reference genes, and PPIA + RPS18 are a suitable reference gene combination. This is a reference gene report that combines transcriptome sequencing analysis and only focuses on ESCC, which makes the quantification more precise, systematic, and standardized, and promotes gene regulation research and the early diagnosis of ESCC in the future.
Collapse
Affiliation(s)
- Jia Xu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Ming Yang
- Department of General Surgery, Fifth People's Hospital of Huaian City, Huaian, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Ai-zhong Shao
- Department of Cardiothorac Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Hui-wen Pan
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yi-xuan Fan
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Ke-ping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
11
|
Švec J, Šťastná M, Janečková L, Hrčkulák D, Vojtěchová M, Onhajzer J, Kříž V, Galušková K, Šloncová E, Kubovčiak J, Pfeiferová L, Hrudka J, Matěj R, Waldauf P, Havlůj L, Kolář M, Kořínek V. TROP2 Represents a Negative Prognostic Factor in Colorectal Adenocarcinoma and Its Expression Is Associated with Features of Epithelial-Mesenchymal Transition and Invasiveness. Cancers (Basel) 2022; 14:4137. [PMID: 36077674 PMCID: PMC9454662 DOI: 10.3390/cancers14174137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/09/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Trophoblastic cell surface antigen 2 (TROP2) is a membrane glycoprotein overexpressed in many solid tumors with a poor prognosis, including intestinal neoplasms. In our study, we show that TROP2 is expressed in preneoplastic lesions, and its expression is maintained in most colorectal cancers (CRC). High TROP2 positivity correlated with lymph node metastases and poor tumor differentiation and was a negative prognostic factor. To investigate the role of TROP2 in intestinal tumors, we analyzed two mouse models with conditional disruption of the adenomatous polyposis coli (Apc) tumor-suppressor gene, human adenocarcinoma samples, patient-derived organoids, and TROP2-deficient tumor cells. We found that Trop2 is produced early after Apc inactivation and its expression is associated with the transcription of genes involved in epithelial-mesenchymal transition, the regulation of migration, invasiveness, and extracellular matrix remodeling. A functionally similar group of genes was also enriched in TROP2-positive cells from human CRC samples. To decipher the driving mechanism of TROP2 expression, we analyzed its promoter. In human cells, this promoter was activated by β-catenin and additionally by the Yes1-associated transcriptional regulator (YAP). The regulation of TROP2 expression by active YAP was verified by YAP knockdown in CRC cells. Our results suggest a possible link between aberrantly activated Wnt/β-catenin signaling, YAP, and TROP2 expression.
Collapse
Affiliation(s)
- Jiří Švec
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
- Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovské Vinohrady, Šrobárova 1150/50, 100 34 Prague, Czech Republic
| | - Monika Šťastná
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Lucie Janečková
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Dušan Hrčkulák
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Martina Vojtěchová
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Jakub Onhajzer
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Vítězslav Kříž
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Kateřina Galušková
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Eva Šloncová
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Lucie Pfeiferová
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
- Department of Informatics and Chemistry, Faculty of Chemical Technology, University of Chemistry and Technology Prague, 166 28 Prague, Czech Republic
| | - Jan Hrudka
- Department of Pathology, Third Faculty of Medicine, Charles University, University Hospital Kralovské Vinohrady, Šrobárova 1150/50, 100 34 Prague, Czech Republic
| | - Radoslav Matěj
- Department of Pathology, Third Faculty of Medicine, Charles University, University Hospital Kralovské Vinohrady, Šrobárova 1150/50, 100 34 Prague, Czech Republic
- Department of Pathology and Molecular Medicine, Third Medical Faculty, Charles University, Thomayer University Hospital, Ruská 87, 100 00 Praha, Czech Republic
| | - Petr Waldauf
- Department of Anaesthesia and Intensive Care Medicine, Third Faculty of Medicine, Charles University, University Hospital Kralovské Vinohrady, Šrobárova 1150/50, 100 34 Prague, Czech Republic
| | - Lukáš Havlůj
- Department of General Surgery, Third Faculty of Medicine, Charles University, University Hospital Kralovské Vinohrady, Šrobárova 1150/50, 100 34 Prague, Czech Republic
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Vladimír Kořínek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
12
|
Giannos P, Triantafyllidis KK, Giannos G, Kechagias KS. SPP1 in infliximab resistant ulcerative colitis and associated colorectal cancer: an analysis of differentially expressed genes. Eur J Gastroenterol Hepatol 2022; 34:598-606. [PMID: 35102110 DOI: 10.1097/meg.0000000000002349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Infliximab, a tumour necrosis factor-α (TNFα) antagonist, has advanced the management of ulcerative colitis. Although efficacious, considerable percentage of patients are resistant to treatment. Accumulative inflammatory burden in long-term ulcerative colitis patients refractory to therapy increases the risk of developing colorectal cancer (CRC). Our study investigated anti-TNFα-naïve patients with active ulcerative colitis to identify gene biomarkers whose dysregulated expression correlated with resistance to infliximab (IFX) treatment and poor prognosis in CRC. METHODS Differentially expressed genes (DEGs) from two studies (GSE73661 and GSE14580) with colonic mucosal samples were retrieved. Noninflammatory bowel disease controls were compared with those with active ulcerative colitis that either responded or were resistant to IFX before treatment. DEGs from ulcerative colitis samples resistant to IFX were used to construct a protein-protein interaction network, and clustering gene modules were identified. Module DEGs that overlapped with ulcerative colitis samples responsive to IFX were analysed, based on topological closeness and radiality. Hub genes were obtained, and their correlation with CRC progression was evaluated. Their expression in CRC tissues and their tumour microenvironment immune status was estimated. RESULTS Three clusters composed of 582 DEGs from ulcerative colitis samples resistant to IFX were retrieved. Comparative analysis identified 305 overlapping DEGs with ulcerative colitis samples responsive to IFX. Topological analysis revealed a hub gene - SPP1 - whose overexpression in CRC tissues and patients correlated with increased infiltration of immune signatures and poor prognosis. CONCLUSION SPP1 may serve as potential gene biomarker and predictor of resistance to IFX therapy in ulcerative colitis and CRC development.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London
- Society of Meta-research and Biomedical Innovation, London
| | | | - Georgios Giannos
- Second Department of Surgery, Evaggelismos Hospital, Athens
- Department of Medicine, University of Crete Medical School, Heraklion, Crete, Greece
| | - Konstantinos S Kechagias
- Society of Meta-research and Biomedical Innovation, London
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London
- Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital National Health Service (NHS) Foundation Trust, London, UK
| |
Collapse
|
13
|
Deepti P, Pasha A, Kumbhakar DV, Doneti R, Heena SK, Bhanoth S, Poleboyina PK, Yadala R, S D A, Pawar SC. Overexpression of Secreted Phosphoprotein 1 (SPP1) predicts poor survival in HPV positive cervical cancer. Gene X 2022; 824:146381. [PMID: 35271951 DOI: 10.1016/j.gene.2022.146381] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/24/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer (CC) is the most prevalent malignant gynecological tumor with limited treatments. The present study describes the role of SPP1 in cancer progression, SPP1 emerged as one of the most overexpressed genes identified through clariom D transcriptome microarray. This investigation aims towards identifying a potential gene with significant prognostic value for detection and early diagnosis of cervical cancer. The elevated expression of SPP1 in cervical squamous cell carcinoma tissue was validated across GEO (Gene Expression Omnibus) microarray data sets, TCGA (The Cancer Genome Atlas), and Oncomine databases. SPP1 expression was found to be prognostically significant, showing association with poor survival rate of the patients. Our study intended to assess the expression of secreted phosphoprotein (SPP1) gene at mRNA and protein levels, and to explore the association of single nucleotide polymorphisms of SPP1 with risk of CC. Further, receiver operating characteristics (ROC) curve was plotted to determine the levels of SPP1 to differentiate CC against control. Results revealed significant (p < 0.01) stage-wise upregulation of SPP1 in CC compared to the normal cervical tissue and this was further confirmed using Immunohistochemistry and real-time PCR. The ROC for SPP1 demonstrated good selective power to differentiate malignant CC and non-malignant cervical tissues. The SPP1 gene -443 T > C promoter polymorphisms are found to be significantly predominant in the disease group and Insilico analysis by the TRANSFAC software confirms its association with loss of STAT6 transcription factor binding site leading to overexpression of the SPP1.
Collapse
Affiliation(s)
- Pasumarthi Deepti
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Akbar Pasha
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Divya Vishambhar Kumbhakar
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Ravinder Doneti
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - S K Heena
- Department of Pathology, Osmania Medical College, Hyderabad, 500095 Telangana, India
| | - Shivaji Bhanoth
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Pavan Kumar Poleboyina
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Rajesh Yadala
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Annapurna S D
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India
| | - Smita C Pawar
- Department of Genetics & Biotechnology, University College of Science, Osmania University, Hyderabad 500007, Telangana, India.
| |
Collapse
|
14
|
Chen QF, Gao H, Pan QY, Wang YJ, Zhong XN. Analysis at the single-cell level indicates an important role of heterogeneous global DNA methylation status on the progression of lung adenocarcinoma. Sci Rep 2021; 11:23337. [PMID: 34857857 PMCID: PMC8639744 DOI: 10.1038/s41598-021-02786-y] [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] [Received: 04/11/2021] [Accepted: 10/15/2021] [Indexed: 12/19/2022] Open
Abstract
Aberrant DNA modifications affect the tumorigenesis and progression of lung cancer. However, the global methylation status of tumor cells and the heterogeneous methylation status of cells within the same tumor need further study. We used publicly available single-cell RNAseq data to investigate the impact and diversity of global methylation status on lung adenocarcinoma. Clustering cells into subgroups and cell differentiation pseudotime analysis, based on expression profile, demonstrated that the global methylation status was crucial to lung adenocarcinoma function and progression. Hypermethylated tumor cells had increased activity related to the hypoxia response. Hyper- and hypomethylated cells indicated upregulation in pathways involving focal adhesion and cell junctions. Pseudotime analysis showed that cell clusters with unique methylation activities were located at the ends of the putative trajectories, suggesting that DNA methylation and demethylation activities were essential to tumor cell progression. Expression of SPP1 was associated with the global methylation status of tumor cells and with patient prognosis. Our study identified the importance and diversity of global DNA methylation status by analysis at the single-cell level. Our findings provide new information about the global DNA methylation status of tumor cells and suggest new approaches for precision medical treatments for lung adenocarcinoma.
Collapse
Affiliation(s)
- Quan-Fang Chen
- Department of Respiratory, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Han Gao
- Department of Respiratory, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Qing-Yun Pan
- Department of Respiratory, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Ying-Ju Wang
- Department of Respiratory, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Xiao-Ning Zhong
- Department of Respiratory, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
15
|
Zhang X, Bi K, Tu X, Zhang Q, Cao Q, Liang Y, Zeng P, Wang L, Liu T, Fang W, Diao H. Interleukin-33 as an early predictor of cetuximab treatment efficacy in patients with colorectal cancer. Cancer Med 2021; 10:8338-8351. [PMID: 34664425 PMCID: PMC8633246 DOI: 10.1002/cam4.4331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/25/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cetuximab is used for colorectal cancer (CRC) treatment. However, the early biomarker of treatment efficacy of cetuximab has not been identified. METHODS After 1 year of cetuximab treatment, patients were divided into an effective group and an ineffective group. The interleukin-33 (IL-33) level and the distribution of lymphatic cells in patients were investigated by analyzing the peripheral blood mononuclear cells via flow cytometry analysis and ELISA. The correlation between IL-33 immunomodulatory effect and cetuximab treatment efficacy was determined through experiments in vivo and in vitro. RESULTS The IL-33 level in the peripheral blood was increased at 4 weeks after cetuximab administration of effective group, meanwhile, the osteopontin (OPN) was reduced. Whereas neither IL-33 level nor OPN level of ineffective patients changed. In the effective group, the number of natural killer (NK) and CD8+ T cells were increased. Moreover, CD137 and CD107a expression on NK cells were higher in the effective group compared to the ineffective group. In vitro cetuximab treatment also increased the number of NK and CD8+ T cells as well as CD137 and CD107a expression upon IL-33 stimulation. Moreover, the secretion of OPN was inhibited by IL-33 administration in cetuximab-treated PBMCs from the effective group patients. IL-33 upregulated the cytotoxicity of NK cells and inhibited tumor cells growth in the effective cetuximab treatment mice. CONCLUSION Effective cetuximab treatment induced a change of IL-33 and OPN at the early stage and triggered the NK cells antitumor activity. Consequently, significantly increased IL-33 level and decreased OPN level in the peripheral blood at the early treatment are proposed as potential predictors of cetuximab treatment efficacy.
Collapse
Affiliation(s)
- Xujun Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Kefan Bi
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Xiaoxuan Tu
- Department of Medical OncologyFirst Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiong Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qingyi Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Yan Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Ping Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lin Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Tianxing Liu
- Department of Biological SciencesUniversity of TorontoTorontoOntarioCanada
| | - Weijia Fang
- Department of Medical OncologyFirst Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
16
|
Zhang H, Wu Y, Li H, Sun L, Meng X. Model constructions of chemosensitivity and prognosis of high grade serous ovarian cancer based on evaluation of immune microenvironment and immune response. Cancer Cell Int 2021; 21:593. [PMID: 34736480 PMCID: PMC8567582 DOI: 10.1186/s12935-021-02295-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/21/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prognosis of high grade serous ovarian cancer (HGSOC) patients is closely related to the immune microenvironment and immune response. Based on this, the purpose of this study was to construct a model to predict chemosensitivity and prognosis, and provide novel biomarkers for immunotherapy and prognosis evaluation of HGSOC. METHODS GSE40595 (38 samples), GSE18520 (63 samples), GSE26712 (195 samples), TCGA (321 samples) and GTEx (88 samples) were integrated to screen differential expressed genes (DEGs) of HGSOC. The prognosis related DEGs (DEPGs) were screened through overall survival analysis. The DEGs-encoded protein-protein interaction network was constructed and hub genes of DEPGs (DEPHGs) were generated by STRING. Immune characteristics of the samples were judged by ssGSEA, ESTIMATE and CYBERSORT. TIMER was used to analyze the relationship between DEPHGs and tumor-infiltrating immunocytes, as well as the immune checkpoint genes, finally immune-related DEPHGs (IDEPHGs) were determined, and whose expression in 12 pairs of HGSOC tissues and tumor-adjacent tissues were analyzed by histological verification. Furthermore, the chemosensitivity genes in IDEPHGs were screened according to GSE15622 (n = 65). Finally, two prediction models of paclitaxel sensitivity score (PTX score) and carboplatin sensitivity score (CBP score) were constructed by lasso algorithm. The area under curve was calculated to estimate the accuracy of candidate gene models in evaluating chemotherapy sensitivity. RESULTS 491 DEGs were screened and 37 DEGs were identified as DEPGs, and 11 DEPHGs were further identified. Among them, CXCL13, IDO1, PI3, SPP1 and TRIM22 were screened as IDEPHGs and verified in the human tissues. Further analysis showed that IDO1, PI3 and TRIM22 could independently affect the chemotherapy sensitivity of HGSOC patients. The PTX score was significantly better than TRIM22, PI3, SPP1, IDO1 and CXCL13 in predicting paclitaxel sensitivity, so was CBP score in predicting carboplatin sensitivity. What's more, both of the HGSOC patients with high PTX score or high CBP score had longer survival time. CONCLUSIONS Five IDEPHGs identified through comprehensive bioinformatics analysis were closely related with the prognosis, immune microenvironment and chemotherapy sensitivity of HGSOC. Two prediction models based on IDEPHGs might have potential application of chemotherapy sensitivity and prognosis for patients with HGSOC.
Collapse
Affiliation(s)
- Han Zhang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yijun Wu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Hao Li
- Department of Clinical Laboratory, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Liping Sun
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, and Key Laboratory of Gastrointestinal Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiangkai Meng
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
17
|
Wang C, Shi Z, Zhang Y, Li M, Zhu J, Huang Z, Zhang J, Chen J. CBFβ promotes colorectal cancer progression through transcriptionally activating OPN, FAM129A, and UPP1 in a RUNX2-dependent manner. Cell Death Differ 2021; 28:3176-3192. [PMID: 34050318 PMCID: PMC8563980 DOI: 10.1038/s41418-021-00810-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is commonly associated with aberrant transcription regulation, but characteristics of the dysregulated transcription factors in CRC pathogenesis remain to be elucidated. In the present study, core-binding factor β (CBFβ) is found to be significantly upregulated in human CRC tissues and correlates with poor survival rate of CRC patients. Mechanistically, CBFβ is found to promote CRC cell proliferation, migration, invasion, and inhibit cell apoptosis in a RUNX2-dependent way. Transcriptome studies reveal that CBFβ and RUNX2 form a transcriptional complex that activates gene expression of OPN, FAM129A, and UPP1. Furthermore, CBFβ significantly promotes CRC tumor growth and live metastasis in a mouse xenograft model and a mouse liver metastasis model. In addition, tumor-suppressive miR-143/145 are found to inhibit CBFβ expression by specifically targeting its 3'-UTR region. Consistently, an inverse correlation between miR-143/miR-145 and CBFβ expression levels is present in CRC patients. Taken together, this study uncovers a novel regulatory role of CBFβ-RUNX2 complex in the transcriptional activation of OPN, FAM129A, and UPP1 during CRC development, and may provide important insights into CRC pathogenesis.
Collapse
Affiliation(s)
- Chen Wang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Ziyu Shi
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Yuqian Zhang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Mingyue Li
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Jie Zhu
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Zhen Huang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Junfeng Zhang
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China
| | - Jiangning Chen
- grid.41156.370000 0001 2314 964XState Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, PR China ,grid.41156.370000 0001 2314 964XState Key Laboratory of Analytical Chemistry for Life Sciences, Nanjing University, Nanjing, PR China
| |
Collapse
|
18
|
Nazarizadeh A, Alizadeh-Fanalou S, Hosseini A, Mirzaei A, Salimi V, Keshipour H, Safizadeh B, Jamshidi K, Bahrabadi M, Tavakoli-Yaraki M. Evaluation of local and circulating osteopontin in malignant and benign primary bone tumors. J Bone Oncol 2021; 29:100377. [PMID: 34235049 PMCID: PMC8246632 DOI: 10.1016/j.jbo.2021.100377] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose The development of novel and efficient biomarkers for primary bone cancers is of grave importance. Methods The expression pattern of osteopontin (OPN) was investigated in the 153 patients with benign (n = 72) and malignant (n = 81) primary bone cancers. Both local and circulating OPN mRNA expression levels and their protein concentration in serum and tumor site were assessed using real-time qRT-PCR, ELISA, and immunohistochemistry techniques, respectively. As a control, 29 healthy individuals were considered. The number of 153 tumor tissue specimens and the 153 paired margins were taken on surgical resection from the patients. 153 blood samples were also drained from all participants, then peripheral blood mononuclear cells (PBMC) and sera were separated. Results The mean mRNA expression was significantly higher in all of the cancerous tissues than the paired margins and the PBMC of the patients than the controls. Consistently, the protein concentrations of OPN in serum and tumor tissues were significantly higher in the patients. Furthermore, the malignant cases had significantly elevated the mRNA levels and the protein compared to the benign cases. OPN could potentially differentiate the patients from the controls with 100% sensitivity and specificity in serum. Moreover, OPN could predict some of the malignant cases' clinicopathological features, including metastasis, recurrence, grade, and response to chemotherapy. Conclusions In conclusion, OPN might be involved in the pathogenesis of primary bone tumors and can be considered as a potential biomarker to bone cancer diagnosis.
Collapse
Key Words
- ANOVA, One-way analysis of variance
- AUC, area under the curve
- Bone tumors
- CI, confidence interval
- Chondrosarcoma
- DAPI, 4′,6-Diamidine-2′-phenylindole dihydrochloride
- ELISA, Enzyme-linked immunosorbent assay
- EMT, epithelial-mesenchymal transition
- Ewing Sarcoma
- HIF-1α, hypoxia-inducible factor-1 alpha
- HRP, horseradish peroxidase
- MMP9, Matrix metallopeptidase 9
- OCT, Optimal Cutting Temperature
- OPN, Osteopontin
- Osteopontin
- Osteosarcoma
- PBMC, Peripheral blood mononuclear cells
- PBS, phosphate-buffered saline
- ROC, receiver operating characteristic
- S100A8, S100 calcium-binding protein A8
- SOX9, SRY-Box Transcription Factor 9
- cDNA, Complementary DNA
- qRT-PCR, Quantitative Real-time transcription-polymerase chain reaction
Collapse
Affiliation(s)
- Ali Nazarizadeh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahin Alizadeh-Fanalou
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ameinh Hosseini
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Mirzaei
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadi Keshipour
- Department of Epidemiology, Faculty of Veterinary Sciences, University of Tehran, Tehran, Iran
| | - Banafsheh Safizadeh
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khodamorad Jamshidi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Bahrabadi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Wei R, Wu Q, Ai N, Wang L, Zhou M, Shaw C, Chen T, Ye RD, Ge W, Siu SW, Kwok HF. A novel bioengineered fragment peptide of Vasostatin-1 exerts smooth muscle pharmacological activities and anti-angiogenic effects via blocking VEGFR signalling pathway. Comput Struct Biotechnol J 2021; 19:2664-2675. [PMID: 34093983 PMCID: PMC8131715 DOI: 10.1016/j.csbj.2021.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
Chromogranin A (CgA) is a hydrophilic glycoprotein released by post-ganglionic sympathetic neurons. CgA consists of a single peptide chain containing numerous paired basic residues, which are typical cleavage sites in prohormones to generate bioactive peptides. It is recognized as a diagnostic and prognostic serum marker for neuroendocrine tumours. Vasostatin-1 is one of the most conserved regions of CgA and has diverse inhibitory biological activities. In this study, a novel peptide fragment that contains three typical functional structures of Vasostatin-1 was synthesized. This unique bioengineered Vasostatin-1 Derived Peptide (named V1DP) includes a highly conserved domain between vertebrate species in its N-terminal region, comprising a disulphide bridge formed by two cysteine residues at amino acid positions 17 and 38, respectively. Besides, V1DP contains two significant tripeptide recognition sequences: the amino acid triplets, RGD and KGD. Our data demonstrated that V1DP could induce a dose-dependent relaxation of rat arterial smooth muscle and also increase the contraction activity of rat uterus smooth muscle. More importantly, we found that V1DP inhibits cancer cell proliferation, modulate the HUVEC cell migration, and exhibit anti-angiogenesis effect both in vitro and in vivo. We further investigated the actual mechanism of V1DP, and our results confirmed that V1DP involves inhibiting the vascular endothelial growth factor receptor (VEGFR) signalling. We docked V1DP to the apo structures of VEGFR2 and examined the stability of the peptide in the protein pockets. Our simulation and free energy calculations results indicated that V1DP can bind to the catalytic domain and regulatory domain pockets, depending on whether the conformational state of the protein is JM-in or JM-out. Taken together, our data suggested that V1DP plays a role as the regulator of endothelial cell function and smooth muscle pharmacological homeostasis. V1DP is a water-soluble and biologically stable peptide and could further develop as an anti-angiogenic drug for cancer treatment.
Collapse
Affiliation(s)
- Ran Wei
- CCZU-JITRI Joint Bio-X Lab, School of Pharmacy & School of Medicine, Changzhou University, Changzhou, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Qiushuang Wu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Nana Ai
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, Avenida de Universidade, University of Macau, Macau SAR, China
| | - Lei Wang
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Northern Ireland, United Kingdom
| | - Mei Zhou
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Northern Ireland, United Kingdom
| | - Chris Shaw
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Northern Ireland, United Kingdom
| | - Tianbao Chen
- Natural Drug Discovery Group, School of Pharmacy, Queen’s University Belfast, Northern Ireland, United Kingdom
| | - Richard Dequan Ye
- School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, Avenida de Universidade, University of Macau, Macau SAR, China
| | - Shirley W.I. Siu
- Department of Computer and Information Science, Faculty of Science and Technology University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Hang Fai Kwok
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
- Corresponding author at: Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China.
| |
Collapse
|
20
|
Yi J, Liu Y, Zhang L, Fang C. Secreted phosphoprotein-1 accelerates the progression of human colorectal cancer through activating β-catenin signaling. Oncol Lett 2021; 21:372. [PMID: 33777196 DOI: 10.3892/ol.2021.12633] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 12/07/2020] [Indexed: 01/05/2023] Open
Abstract
Colorectal cancer (CRC) is a common malignant tumor of the digestive tract and one of the leading causes of cancer-associated mortality. Secreted phosphoprotein-1 (SPP-1) is overexpressed in CRC and promotes cancer progression, but the underlying mechanisms underlying SPP-1 function remain unclear. The present study aimed to explore the effects of Wnt/β-catenin signaling in SPP-1-induced CRC progression. The expression patterns of SPP-1 in CRC tissues were examined using reverse transcription-quantitative (RT-q)PCR, western blotting and immunohistochemistry. SPP-1 expression in cells was assessed using RT-qPCR and western blotting. Cell-Counting Kit-8, flow cytometry and tumor-burdened mice experiments were used to determine cell proliferation, apoptosis and in vivo tumor formation abilities. The results showed that SPP-1 expression was markedly elevated in CRC tissues and cells compared with that in normal colorectal tissues and cells. High expression of SPP-1 was associated with advanced clinical process and low overall survival rate in patients with CRC. Besides, SPP-1 could interact with β-catenin and positively regulated β-catenin protein expression, and enhanced its nuclear accumulation. Moreover, SPP-1-upregulation significantly enhanced cell proliferation and in vivo tumor formation ability, and reduced apoptosis, whereas these effects were all abolished when β-catenin was silenced. Overall, the present study revealed that SPP-1 promoted the progression of CRC in a β-catenin-dependent manner.
Collapse
Affiliation(s)
- Jianzhong Yi
- Department of Gastroenteric Hernia, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Yungeng Liu
- Department of Gastroenteric Hernia, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Lei Zhang
- Department of Gastroenteric Hernia, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, P.R. China
| | - Chuanfa Fang
- Department of Gastroenteric Hernia, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
21
|
Stromal Protein-Mediated Immune Regulation in Digestive Cancers. Cancers (Basel) 2021; 13:cancers13010146. [PMID: 33466303 PMCID: PMC7795083 DOI: 10.3390/cancers13010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Solid cancers are surrounded by a network of non-cancerous cells comprising different cell types, including fibroblasts, and acellular protein structures. This entire network is called the tumor microenvironment (TME) and it provides a physical barrier to the tumor shielding it from infiltrating immune cells, such as lymphocytes, or therapeutic agents. In addition, the TME has been shown to dampen efficient immune responses of infiltrated immune cells, which are key in eliminating cancer cells from the organism. In this review, we will discuss how TME proteins in particular are involved in this dampening effect, known as immunosuppression. We will focus on three different types of digestive cancers: pancreatic cancer, colorectal cancer, and gastric cancer. Moreover, we will discuss current therapeutic approaches using TME proteins as targets to reverse their immunosuppressive effects. Abstract The stromal tumor microenvironment (TME) consists of immune cells, vascular and neural structures, cancer-associated fibroblasts (CAFs), as well as extracellular matrix (ECM), and favors immune escape mechanisms promoting the initiation and progression of digestive cancers. Numerous ECM proteins released by stromal and tumor cells are crucial in providing physical rigidity to the TME, though they are also key regulators of the immune response against cancer cells by interacting directly with immune cells or engaging with immune regulatory molecules. Here, we discuss current knowledge of stromal proteins in digestive cancers including pancreatic cancer, colorectal cancer, and gastric cancer, focusing on their functions in inhibiting tumor immunity and enabling drug resistance. Moreover, we will discuss the implication of stromal proteins as therapeutic targets to unleash efficient immunotherapy-based treatments.
Collapse
|
22
|
Kwok H. Meet Our Editorial Board Member. Curr Protein Pept Sci 2021. [DOI: 10.2174/138920372201210301112500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- H.F. Kwok
- University of Macau Taipa,Macao,China
| |
Collapse
|
23
|
Yang C, Pan H, Shen L. Pan-Cancer Analyses Reveal Prognostic Value of Osteomimicry Across 20 Solid Cancer Types. Front Mol Biosci 2020; 7:576269. [PMID: 33240930 PMCID: PMC7678014 DOI: 10.3389/fmolb.2020.576269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/12/2020] [Indexed: 11/16/2022] Open
Abstract
Background Osteomimicry of cancer cells had been widely reported in prostate cancer and breast cancer. However, the prognostic value of osteomimicry in various cancer types remained unclear. We hypothesized that osteomimicry would result in remodeling of the tumor microenvironment and was eligible to predict patient prognosis. Methods A comprehensive transcriptomic analysis of the osteomimicry, which was characterized by mRNA expression of SPARC, SPP1, and BGLAP, across 20 solid tumors (7564 patients) using RNA-seq data from The Cancer Genome Atlas (TCGA) was conducted. Samples of each cancer type were classified into subgroups (high vs. low) based on median value of osteomimetic markers, the associations of these markers with clinical outcomes, immune cell infiltration and immune checkpoints expression were explored. Results Each osteomimetic marker harbored prognostic value in the pan-cancer analyses [SPARC: hazard ratio (HR) = 1.10, p = 0.028; SPP1: HR = 1.25, p < 0.001; BGLAP: HR = 1.13, p = 0.005]. Patients with high expression of all the three genes also had significantly unfavorable survival (HR = 1.61, p < 0.0001) compared with those of low expression. Correlation analyses demonstrated that osteomimicry was closely related to tumor purity, dendritic cells (DC) infiltration and expression of immune checkpoints. Conclusion Osteomimicry had prognostic value in various cancer types and the underlying mechanism might correlate to the trapping and dysfunction of DCs in the tumor microenvironment, revealing the potential of osteomimicry as a target of immunotherapy.
Collapse
Affiliation(s)
- Changsheng Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Southern Medical University, Orthopaedic Hospital of Guangdong Province, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Southern Medical University, Guangzhou, China
| | - Hehai Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lujun Shen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
24
|
Wei T, Bi G, Bian Y, Ruan S, Yuan G, Xie H, Zhao M, Shen R, Zhu Y, Wang Q, Yang Y, Zhu D. The Significance of Secreted Phosphoprotein 1 in Multiple Human Cancers. Front Mol Biosci 2020; 7:565383. [PMID: 33324676 PMCID: PMC7724571 DOI: 10.3389/fmolb.2020.565383] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/24/2020] [Indexed: 12/12/2022] Open
Abstract
Malignant tumor represents a major reason for death in the world and its incidence is growing rapidly. Developing the tools for early diagnosis is possibly a promising way to offer diverse therapeutic options and promote the survival chance. Secreted phosphoprotein 1 (SPP1), also called Osteopontin (OPN), has been demonstrated overexpressed in many cancers. However, the specific role of SPP1 in prognosis, gene mutations, and changes in gene and miRNA expression in human cancers is unclear. In this report, we found SPP1 expression was higher in most of the human cancers. Based on Kaplan-Meier plotter and the PrognoScan database, we found high SPP1 expression was significantly correlated with poor survival in various cancers. Using a large dataset of colon adenocarcinoma (COAD), head and neck cancer (HNSC), lung adenocarcinoma (LUAD), and lung squamous cell carcinoma (LUSC) patients from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, this study identified 22 common genes and 2 common miRNAs. GO, and KEGG paths analyses suggested that SPP1 correlated genes were mainly involved in positive regulation of immune cell activation and infiltration. SPP1-associated genes and miRNAs regulatory networks suggested that their interactions may play a role in the progression of four selected cancers. SPP1 showed significant positive correlation with the immunocyte and immune marker sets infiltrating degrees. All of these data provide strong evidence that SPP1 may promote tumor progress through interacting with carcinogenic genes and facilitating immune cells’ infiltration in COAD, HNSC, LUAD, and LUSC.
Collapse
Affiliation(s)
- Tengteng Wei
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suhong Ruan
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Guangda Yuan
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Hongya Xie
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongming Shen
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Yimeng Zhu
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Yang
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Donglin Zhu
- Department of Thoracic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
25
|
Osteopontin: A Key Regulator of Tumor Progression and Immunomodulation. Cancers (Basel) 2020; 12:cancers12113379. [PMID: 33203146 PMCID: PMC7698217 DOI: 10.3390/cancers12113379] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Anti-PD-1/PD-L1 and anti-CTLA-4-based immune checkpoint blockade (ICB) immunotherapy have recently emerged as a breakthrough in human cancer treatment. Durable efficacy has been achieved in many types of human cancers. However, not all human cancers respond to current ICB immunotherapy and only a fraction of the responsive cancers exhibit efficacy. Osteopontin (OPN) expression is highly elevated in human cancers and functions as a tumor promoter. Emerging data suggest that OPN may also regulate immune cell function in the tumor microenvironment. This review aims at OPN function in human cancer progression and new findings of OPN as a new immune checkpoint. We propose that OPN compensates PD-L1 function to promote tumor immune evasion, which may underlie human cancer non-response to current ICB immunotherapy. Abstract OPN is a multifunctional phosphoglycoprotein expressed in a wide range of cells, including osteoclasts, osteoblasts, neurons, epithelial cells, T, B, NK, NK T, myeloid, and innate lymphoid cells. OPN plays an important role in diverse biological processes and is implicated in multiple diseases such as cardiovascular, diabetes, kidney, proinflammatory, fibrosis, nephrolithiasis, wound healing, and cancer. In cancer patients, overexpressed OPN is often detected in the tumor microenvironment and elevated serum OPN level is correlated with poor prognosis. Initially identified in activated T cells and termed as early T cell activation gene, OPN links innate cells to adaptive cells in immune response to infection and cancer. Recent single cell RNA sequencing revealed that OPN is primarily expressed in tumor cells and tumor-infiltrating myeloid cells in human cancer patients. Emerging experimental data reveal a key role of OPN is tumor immune evasion through regulating macrophage polarization, recruitment, and inhibition of T cell activation in the tumor microenvironment. Therefore, in addition to its well-established direct tumor cell promotion function, OPN also acts as an immune checkpoint to negatively regulate T cell activation. The OPN protein level is highly elevated in peripheral blood of human cancer patients. OPN blockade immunotherapy with OPN neutralization monoclonal antibodies (mAbs) thus represents an attractive approach in human cancer immunotherapy.
Collapse
|
26
|
Dos Santos ES, Ramos JC, Roza ALOC, Mariz BALA, Paes Leme AF. The role of osteopontin in oral cancer: A brief review with emphasis on clinical applications. Oral Dis 2020; 28:326-335. [PMID: 33188646 DOI: 10.1111/odi.13716] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/13/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Osteopontin (OPN) is a calcium-binding glycol-phosphoprotein present in many physiologic and pathological processes. This protein can control bone cell adhesion, osteoclastic activity, and bone matrix mineralization. However, its participation in pathological processes such as atherosclerosis, sarcoidosis, tuberculosis, and cancer have been described. Some studies have shown that OPN may participate in the development and progression of oral cancer. Although the role of OPN in oral cancer is not fully understood, some studies have suggested that this protein may induce malignant phenotype of cells by activation of PI3K/AKT/mTOR pathway, which favors cell proliferation, invasion, metastasis, angiogenesis, and failure of treatment. This review discusses the possible mechanism of involvement of OPN in oral cancer and its potential clinical application in diagnosis and prognosis.
Collapse
Affiliation(s)
| | - Joab Cabral Ramos
- Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | | | | | - Adriana Franco Paes Leme
- Brazilian Bioscience National Laboratory, Brazil Center of Research in Energy and Materials, Campinas, Brazil
| |
Collapse
|
27
|
Liu Y, Li C, Dong L, Chen X, Fan R. Identification and verification of three key genes associated with survival and prognosis of COAD patients via integrated bioinformatics analysis. Biosci Rep 2020; 40:BSR20200141. [PMID: 32936304 PMCID: PMC7522359 DOI: 10.1042/bsr20200141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 08/21/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most lethal malignancy in the world, wherein colon adenocarcinoma (COAD) is the most prevalent type of CRC. Exploring biomarkers is important for the diagnosis, treatment, and prevention of COAD. METHODS We used GEO2R and Venn online software for differential gene screening analysis. Hub genes were screened via Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and Cytoscape, following Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Finally, survival analysis and RNA expression validation were performed via UALCAN online software and real-time PCR. Immunohistochemistry (IHC) was performed to verify the protein expression level of hub genes from tissues of COAD patients. RESULTS In the present study, we screened 323 common differentially expressed genes (DEGs) from four GSE datasets. Furthermore, four hub genes were selected for survival correlation analysis and expression level verification, three of which were shown to be statistically significant. CONCLUSION Our study suggests that Serpin Family E Member 1 (SERPINE1), secreted phosphoprotein 1 (SPP1) and tissue inhibitor of metalloproteinase 1 (TIMP1) may be biomarkers closely related to the prognosis of CRC patients.
Collapse
Affiliation(s)
- Yong Liu
- Department of General Surgery, Tianjin Xiqing Hospital, Tianjin 300380, P.R. China
| | - Chao Li
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, P.R. China
| | - Lijin Dong
- Editorial Department of Education and Research Security Centre, Logistic University of Chinese People’s Armed Police Force, Tianjin 300309, P.R. China
| | - Xuewei Chen
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, P.R. China
| | - Rong Fan
- Department of Operational Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, P.R. China
- Central Laboratory, Tianjin Xiqing Hospital, Tianjin 300380, P.R. China
| |
Collapse
|
28
|
Gerarduzzi C, Hartmann U, Leask A, Drobetsky E. The Matrix Revolution: Matricellular Proteins and Restructuring of the Cancer Microenvironment. Cancer Res 2020; 80:2705-2717. [PMID: 32193287 DOI: 10.1158/0008-5472.can-18-2098] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2019] [Accepted: 03/17/2020] [Indexed: 11/16/2022]
Abstract
The extracellular matrix (ECM) surrounding cells is indispensable for regulating their behavior. The dynamics of ECM signaling are tightly controlled throughout growth and development. During tissue remodeling, matricellular proteins (MCP) are secreted into the ECM. These factors do not serve classical structural roles, but rather regulate matrix proteins and cell-matrix interactions to influence normal cellular functions. In the tumor microenvironment, it is becoming increasingly clear that aberrantly expressed MCPs can support multiple hallmarks of carcinogenesis by interacting with various cellular components that are coupled to an array of downstream signals. Moreover, MCPs also reorganize the biomechanical properties of the ECM to accommodate metastasis and tumor colonization. This realization is stimulating new research on MCPs as reliable and accessible biomarkers in cancer, as well as effective and selective therapeutic targets.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada. .,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Elliot Drobetsky
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
29
|
Tu Y, Chen C, Fan G. Association between the expression of secreted phosphoprotein - related genes and prognosis of human cancer. BMC Cancer 2019; 19:1230. [PMID: 31849319 PMCID: PMC6918603 DOI: 10.1186/s12885-019-6441-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 12/08/2019] [Indexed: 12/15/2022] Open
Abstract
Background While many studies have assessed the predictive value of secreted phosphoprotein (SPP) genes in cancer, the findings have been inconsistent. To resolve these inconsistencies, we systematically analyzed the available data to determine whether SPP1 and SPP2 are prognostic markers in the context of human cancer. Methods The expression of SPP1 and SPP2 was assessed by Oncomine analysis. The PrognoScan database was used to assess the prognostic value of SPP1 and SPP2, with cBioPortal used to assess copy number variations. The STRING database was used to generate a Protein - Protein Interaction (PPI) network for SPP genes. Results SPP1 was more likely to be over-expressed in breast, bladder, colorectal, head, neck, liver, lung, and esophageal cancers. SPP2 was expressed at lower levels in colorectal cancer, leukemia, liver cancer and pancreatic cancer. In addition, SPP1 and SPP2 mutations mainly occurred in cutaneous melanoma and endometrial cancer. Conclusions Our results suggest that SPP1 and SPP2 may be effective therapeutic or diagnostic targets in certain cancers. Further research is required to confirm these results and verify the value of SPP1 and SPP2 as clinical markers of cancer prognosis.
Collapse
Affiliation(s)
- Yaqin Tu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cai Chen
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guorun Fan
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
30
|
Hao C, Cui Y, Chang S, Huang J, Birkin E, Hu M, Zhi X, Li W, Zhang L, Cheng S, Jiang WG. OPN promotes the aggressiveness of non-small-cell lung cancer cells through the activation of the RON tyrosine kinase. Sci Rep 2019; 9:18101. [PMID: 31792339 PMCID: PMC6889187 DOI: 10.1038/s41598-019-54843-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Osteopontin (OPN) is identified as a diagnostic and prognostic biomarker of tumor progression and metastasis. In non-small-cell lung cancer (NSCLC), the functions of OPN have not been well characterized. The current study sought to investigate the clinical implications of OPN expression in NSCLC and the role of OPN in the aggressiveness of the lung cancer cells. Using a proteomics approach, we identified that phospho-RON (p-RON) was one of the most remarkably up-regulated proteins in OPN-overexpressing cells. The levels of OPN and RON transcripts were unveiled as independent prognostic indicators of survival in NSCLC (p = 0.001). Higher levels of OPN, RON and p-RON proteins were observed in tumor tissues. Knock down of the OPN gene suppressed the migration and invasion abilities of the A549 lung cancer cells which endogenously expresses OPN. While ectopic expression of OPN in the SK-MES-1 lung cancer cells increased levels of cellular invasion and migration. In addition, these changes were accompanied by a phosphorylated activation of RON. Small-molecule inhibition of RON or siRNA silencing of RON significantly reduced OPN-induced migration and invasion of lung cancer cells and had an inhibitory effect on the OPN-mediated cell epithelial-mesenchymal transition. Our study suggests that in NSCLC, the aberrant expression of OPN can be considered as an independent survival indicator and is associated with disease progression. OPN plays a crucial role in promoting migration and invasion properties of lung cancer cells through its phosphorylation activation of the RON signaling pathway, implying its potential as a therapeutic target in the treatment of NSCLC.
Collapse
Affiliation(s)
- Chengcheng Hao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, 100069, China
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yuxin Cui
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Siyuan Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, 100069, China
| | - Jing Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, 100069, China
| | - Emily Birkin
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Mu Hu
- Department of Thoracic Surgery, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiuyi Zhi
- Department of Thoracic Surgery, Beijing Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Lijian Zhang
- Department of Thoracic Surgery, Peking University School of Oncology and Beijing Cancer Hospital & Institute, Beijing, 100142, P.R. China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, 100069, China.
| | - Wen G Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Cancer & Metastasis Research, Capital Medical University, Beijing, 100069, China.
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
31
|
Assidi M, Gomaa W, Jafri M, Hanbazazh M, Al-Ahwal M, Pushparaj P, Al-Harbi A, Al-Qahtani M, Buhmeida A, Al-Maghrabi J. Prognostic value of Osteopontin (SPP1) in colorectal carcinoma requires a personalized molecular approach. Tumour Biol 2019; 41:1010428319863627. [PMID: 31500540 DOI: 10.1177/1010428319863627] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Stratification of colorectal cancer for better management and tangible clinical outcomes is lacking in clinical practice. To reach this goal, the identification of reliable biomarker(s) is a prerequisite to deliver personalized colorectal cancer theranostics. Osteopontin (SPP1) is a key extracellular matrix protein involved in several pathophysiological processes including cancer progression and metastasis. However, the exact molecular mechanisms regulating its expression, localization, and molecular functions in cancer are still poorly understood. This study was designed to investigate the SPP1 expression profiles in Saudi colorectal cancer patients, and to assess its prognostic value. Hundred thirty-four (134) archival paraffin blocks of colorectal cancer were collected from King Abdulaziz University Hospital, Saudi Arabia. Tissue microarrays were constructed, and automated immunohistochemistry was performed to evaluate SPP1 protein expression patterns in colorectal cancer. About 20% and 23% of our colorectal cancer samples showed high SPP1 cytoplasmic and nuclear expression patterns, respectively. Cytoplasmic SPP1 did not correlate with age, gender, tumor size, and location. However, significant correlations were observed with tumor grade (p = 0.008), tumor invasion (p = 0.01), and distant metastasis (p = 0.04). Kaplan-Meier survival analysis showed a significantly lower recurrence rate in patients with higher SPP1 cytoplasmic expression (p = 0.05). At multivariate analysis, high SPP1 cytoplasmic expression was an independent favorable prognostic marker (p = 0.02). However, nuclear SPP1 expression did not show any prognostic value (p = 0.712). Our results showed a particular SPP1 prognostic relevance that is not in line with most colorectal cancer previous studies that may be attributed to the molecular pathophysiology of our colorectal cancer cohort. Saudi Arabia has both specific genomic makeup and particular environment that could lead to distinctive molecular roots of cancer. SPP1 has several isoforms, tissue localizations and molecular functions, signaling pathways, and downstream molecular functions. Therefore, a more individualized approach for CRC studies and particularly SPP1 prognosis outcomes' assessment is highly recommended toward precision oncology.
Collapse
Affiliation(s)
- Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Medical Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wafaey Gomaa
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pathology, Faculty of Medicine, Minia University, Al Minia, Egypt
| | - Mohammad Jafri
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Medical Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mehenaz Hanbazazh
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud Al-Ahwal
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Medical Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Asia Al-Harbi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelbaset Buhmeida
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jaudah Al-Maghrabi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Jeddah, Saudi Arabia
| |
Collapse
|
32
|
Xu X, Jiang X, Chen L, Zhao Y, Huang Z, Zhou H, Shi M. MiR-181a Promotes Apoptosis and Reduces Cisplatin Resistance by Inhibiting Osteopontin in Cervical Cancer Cells. Cancer Biother Radiopharm 2019; 34:559-565. [PMID: 31436472 DOI: 10.1089/cbr.2019.2858] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: In this study, the authors established a cervical cancer cisplatin (DDP) drug-resistant cell line to explore the role of miR-181a in the regulation of osteopontin (OPN) expression and the proliferation, apoptosis, as well as DDP resistance of cervical cancer cells. Materials and Methods: Dual luciferase reporter gene assay was performed to validate the targeted relationship between miR-181a and OPN. The DDP-resistant cell line CaSki/DDP was established to compare the expressions of miR-181a and OPN. The cell proliferation activity was detected by CCK-8 assay. CaSki/DDP cells were divided into miR-NC group and miR-181a mimic group followed by analysis of cell apoptosis by flow cytometry, and the cell proliferation by EdU staining. Results: There was a targeted relationship between miR-181a and OPN mRNA. MiR-181a expression was significantly lower, while OPN mRNA and protein levels were significantly higher in CaSki/DDP cells than that in CaSki cells. Compared with the miR-NC group, OPN mRNA and protein were significantly decreased, cell apoptosis was significantly increased, and cell proliferation ability was significantly attenuated in miR-181a mimic transfection group. Conclusions: The decrease of miR-181a expression and the upregulation of OPN expression are related to the DDP resistance of cervical cancer cells. Overexpression of miR-181a can inhibit the expression of OPN, induce cell apoptosis cells, restrain cell proliferation, and reduce DDP resistance in cervical cancer cells.
Collapse
Affiliation(s)
- Xiaofei Xu
- Department of Obstetrics and Gynecology, Lishui Hospital of Traditional Chinese Medicine, Lishui, Zhejiang, China
| | - Xiaofei Jiang
- Department of Gynecology, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu, China
| | - Liping Chen
- Department of Cardiology, Lishui Hospital of Traditional Chinese Medicine, Lishui, Zhejiang, China
| | - Yu Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhihua Huang
- Department of Obstetrics and Gynecology, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Huifang Zhou
- Department of Gynecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mingqing Shi
- Department of Obstetrics and Gynecology, Lishui Hospital of Traditional Chinese Medicine, Lishui, Zhejiang, China
| |
Collapse
|
33
|
Cheng Y, Wen G, Sun Y, Shen Y, Zeng Y, Du M, Zhu G, Wang G, Meng X. Osteopontin Promotes Colorectal Cancer Cell Invasion and the Stem Cell-Like Properties through the PI3K-AKT-GSK/3β-β/Catenin Pathway. Med Sci Monit 2019; 25:3014-3025. [PMID: 31017126 PMCID: PMC6496974 DOI: 10.12659/msm.913185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 12/27/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteopontin (OPN) is a molecule expressed in numerous cancers including colorectal cancer (CRC) that correlates disease progression. The interaction of OPN that promotes CRC cell migration, invasion, and cancer stem-like cells (CSCs) have not been elucidated. Hence, we aimed to investigate the mechanisms that might be involved. MATERIAL AND METHODS Expression of OPN in tumor tissues derived from patients was monitored with real-time quantitative polymerase chain reaction and western blot. Wound healing and Transwell assay were used to test the differences in migration and invasion in an OPN enriched environment and OPN knockdown condition. Aldehyde dehydrogenase 1 (ALDH1) positive stem cells were isolated using fluorescence-activated cell sorting (FACS) following the protocol of the ALDEFLUOR™ kit. The expression of protein participation in the PI3K-Akt-GSK/3ß-ß/catenin pathway was detected by western blot. RESULTS OPN exhibited increased levels in CRC tumor tissue compared with non-tumor normal tissue and the high level of which correlated with lymphatic metastasis and late TNM stage. Additional rhOPN co-cultured low-expression CRC cells demonstrated more aggressive capability of proliferation, migration, and invasion. For knockdown of OPN in high-expression CRC cells, the bioactivities of proliferation, migration, and invasion were significantly inhibited. Interestingly, the percentage of ALDH1 labeled stem cells was dramatically decreased by OPN inhibition. The phosphorylation of PI3K-Akt-GSK/3ß-ß/catenin pathway was involved in the OPN signaling. Furthermore, Ly294002, a specific PI3K inhibitor, can reverse the promotion of bioactivities and stem cell proportion among rhOPN treated CRC cells. CONCLUSIONS OPN promoted cell proliferation, migration, and invasion, and was accompanied by upregulation of ALDH1-positive CSC in CRC through activation of PI3K-Akt-GSK/3ß-ß/catenin pathway.
Collapse
Affiliation(s)
- Yuanguang Cheng
- Department of Gastrointestinal Surgery, The First Afliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Gang Wen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yong Sun
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yang Shen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yongqing Zeng
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Ming Du
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Guangyu Zhu
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Guanglong Wang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Xiangling Meng
- Department of Gastrointestinal Surgery, The First Afliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
34
|
Identification of plasma secreted phosphoprotein 1 as a novel biomarker for upper tract urothelial carcinomas. Biomed Pharmacother 2019; 113:108744. [PMID: 30844659 DOI: 10.1016/j.biopha.2019.108744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/25/2019] [Indexed: 12/24/2022] Open
Abstract
The key prognostic factor at the time of diagnosis of upper tract urothelial carcinomas (UTUC) is whether the tumor is in the muscle-invasive or non-muscle invasive stage. It is critical to identify novel molecular biomarkers for early detection and target therapy. Plasma proteins secreted by tumor tissues have excellent potential as biomarkers for UTUC. In this study, we conducted a systematic study to identify plasma markers for UTUC based on RNA-seq data from five UTUC tissues and paired adjacent noncancerous mucosa. Through bioinformatics analysis, we found secreted phosphoprotein 1 (SPP1) was the most significant gene that coding secretory protein. Then, qRT-PCR and enzyme-linked immunosorbent assay were performed to evaluate the expression and clinical significance of SPP1 in UTUC. Results found that SPP1 mRNA was upregulated in UTUC cells and tissues, and high SPP1 mRNA expression level was closely related to advanced stage and high grade. Moreover, it is suggested that plasma SPP1 may be a potential biomarker to help identify early-stage UTUC patients and predict invasive and high-grade UTUC. In conclusion, plasma SPP1 is a novel biomarker for UTUC.
Collapse
|
35
|
Wei R, Wong JPC, Lyu P, Xi X, Tong O, Zhang SD, Yuen HF, Shirasawa S, Kwok HF. In vitro and clinical data analysis of Osteopontin as a prognostic indicator in colorectal cancer. J Cell Mol Med 2018; 22:4097-4105. [PMID: 29851214 PMCID: PMC6111822 DOI: 10.1111/jcmm.13686] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/21/2018] [Indexed: 12/25/2022] Open
Abstract
Osteopontin (OPN) has been shown to promote colorectal cancer (CRC) progression; however, the mechanism of OPN-induced CRC progression is largely unknown. In this study, we found that OPN overexpression led to enhanced anchorage-independent growth, cell migration and invasion in KRAS gene mutant cells but to a lesser extent in KRAS wild-type cells. OPN overexpression also induced PI3K signalling, expression of Snail and Matrix metallopeptidase 9 (MMP9), and suppressed the expression of E-cadherin in KRAS mutant cells. In human CRC specimens, a high-level expression of OPN significantly predicted poorer survival in CRC patients and OPN expression was positively correlated with MMP9 expression, and negatively correlated with E-cadherin expression. Furthermore, we have found that 15 genes were co-upregulated in OPN highly expression CRC and a list of candidate drugs that may have potential to reverse the secreted phosphoprotein 1 (SPP1) gene signature by connectivity mapping. In summary, OPN is a potential prognostic indicator and therapeutic target for colon cancer.
Collapse
Affiliation(s)
- Ran Wei
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | | | - Peng Lyu
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Xinping Xi
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Olivia Tong
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, Londonderry, UK
| | - Hiu Fung Yuen
- Institute of Molecular and Cell Biology, A*STAR, Singapore City, Singapore
| | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| |
Collapse
|