1
|
Samaha J, Madhu S, Shehadeh LA, Martinez CA. Osteopontin as a potential mediator of inflammation in HIV and comorbid conditions. AIDS 2025; 39:483-495. [PMID: 40080169 DOI: 10.1097/qad.0000000000004112] [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: 09/27/2024] [Accepted: 12/23/2024] [Indexed: 03/15/2025]
Abstract
INTRODUCTION Approximately 39 million people live with HIV globally, with 1.3 million new infections annually. Despite improved treatment, noncommunicable diseases (NCDs) such as cardiovascular disease (CVD), neurological disorders, chronic kidney disease (CKD), and cancer are now the leading causes of death among people with HIV (PWH). Osteopontin (OPN) has emerged as a notable mediator in the inflammatory response to HIV and related NCDs. Our aim is to review the current understanding of OPN's role in HIV-related inflammatory pathways to highlight potential therapeutic avenues for improved treatment and mitigation of comorbidities. METHODS We conducted a systematic review by searching relevant literature using specific keywords related to HIV, osteopontin, cardiovascular disease, inflammation, neurological disorders, cancer, and chronic kidney disease. The collected studies were organized and categorized by key themes, followed by a comprehensive analysis to identify patterns and draw conclusions regarding OPN's role in HIV-associated comorbidities. RESULTS The intricate interactions between OPN, its isoforms, and HIV-related illnesses suggest that OPN can exhibit both pro-inflammatory and anti-inflammatory roles, depending on the stage of the disease and the specific cell type involved. Its functions are diverse throughout the progression of HIV and its associated comorbidities, including CVD, CKD, cancer, and neurological disorders. CONCLUSION OPN's effects on the disease progression of HIV and related NCDs are highly variable due to its diverse functions. Therefore, further research is essential to fully understand its complex roles before considering OPN as a therapeutic target for HIV and its comorbidities.
Collapse
Affiliation(s)
- Jacklyn Samaha
- Department of Medicine
- Department of Public Health Sciences, University of Miami Miller School of Medicine
| | - Shashank Madhu
- Department of Medicine
- Department of Public Health Sciences, University of Miami Miller School of Medicine
| | - Lina A Shehadeh
- Department of Medicine
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | | |
Collapse
|
2
|
Xu X, Gan J, Gao Z, Li R, Huang D, Lin L, Luo Y, Yang Q, Xu J, Li Y, Fang Q, Peng T, Wang Y, Xu Z, Huang A, Hong H, Lei F, Huang W, Leng J, Li T, Bo X, Chen H, Li C, Gu J. 3D genome landscape of primary and metastatic colorectal carcinoma reveals the regulatory mechanism of tumorigenic and metastatic gene expression. Commun Biol 2025; 8:365. [PMID: 40038385 PMCID: PMC11880527 DOI: 10.1038/s42003-025-07647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Colorectal carcinoma (CRC) is a deadly cancer with an aggressive nature, and how CRC tumor cells manage to translocate and proliferate in a new tissue environment remains not fully understood. Recently, higher-order chromatin structures and spatial genome organization are increasingly implicated in diseases including cancer, but in-depth studies of three-dimensional genome (3D genome) of metastatic cancer are currently lacking, preventing the understanding of the roles of genome organization during metastasis. Here we perform multi-omics profiling of matched normal colon, primary tumor, lymph node metastasis, liver metastasis and normal liver tissue from CRC patients using Hi-C, ATAC-seq and RNA-seq technologies. We find that widespread alteration of 3D chromatin structure is accompanied by dysregulation of genes including SPP1 during the tumorigenesis or metastasis of CRC. Remarkably, the hierarchy of topological associating domain (TAD) changes dynamically, which challenges the traditional view that the TAD structure between tumor and normal tissue is conservative. In addition, we define compartment stability score to measure large-scale alteration in metastatic tumors. To integrate multi-omics data and recognize candidate genes driving cancer metastasis, a pipeline is developed based on Hi-C, RNA-seq and ATAC-seq data. And three candidate genes ARL4C, FLNA, and RGCC are validated to be associated with CRC cell migration and invasion using in vitro knockout experiments. Overall, these data resources and results offer new insights into the involvement of 3D genome in cancer metastasis.
Collapse
Affiliation(s)
- Xiang Xu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Academy of Military Medical Sciences, Beijing, China
| | - Jingbo Gan
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Center for Precision Diagnosis and Treatment of Colorectal Carcinoma and Inflammatory Diseases, Peking University Health Science Center, Beijing, China
| | - Ruifeng Li
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Dandan Huang
- Center for Precision Diagnosis and Treatment of Colorectal Carcinoma and Inflammatory Diseases, Peking University Health Science Center, Beijing, China
- Department of Oncology, Peking University Shougang Hospital, Beijing, China
| | - Lin Lin
- Academy of Military Medical Sciences, Beijing, China
| | - Yawen Luo
- Academy of Military Medical Sciences, Beijing, China
| | - Qian Yang
- Academy of Military Medical Sciences, Beijing, China
| | - Jingxuan Xu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
| | - Yaru Li
- Academy of Military Medical Sciences, Beijing, China
| | - Qing Fang
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Ting Peng
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Yaqi Wang
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - Zihan Xu
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
| | - An Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Haopeng Hong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Fuming Lei
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Center for Precision Diagnosis and Treatment of Colorectal Carcinoma and Inflammatory Diseases, Peking University Health Science Center, Beijing, China
| | - Wensheng Huang
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China
- Center for Precision Diagnosis and Treatment of Colorectal Carcinoma and Inflammatory Diseases, Peking University Health Science Center, Beijing, China
| | - Jianjun Leng
- Center for Precision Diagnosis and Treatment of Colorectal Carcinoma and Inflammatory Diseases, Peking University Health Science Center, Beijing, China
- Department of Hepatopancreatobiliary Surgery, Peking University Shougang Hospital, Beijing, China
| | - Tingting Li
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Xiaochen Bo
- Academy of Military Medical Sciences, Beijing, China
| | - Hebing Chen
- Academy of Military Medical Sciences, Beijing, China.
| | - Cheng Li
- Center for Bioinformatics, School of Life Sciences, Center for Statistical Science, Peking University, Beijing, China.
| | - Jin Gu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, Beijing, China.
- Center for Precision Diagnosis and Treatment of Colorectal Carcinoma and Inflammatory Diseases, Peking University Health Science Center, Beijing, China.
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- Peking University International Cancer Institute, Beijing, China.
| |
Collapse
|
3
|
Sun J, Tang M, Cai Z. SPP1 promotes tumor progression in esophageal carcinoma by activating focal adhesion pathway. J Gastrointest Oncol 2024; 15:818-828. [PMID: 38989403 PMCID: PMC11231845 DOI: 10.21037/jgo-24-302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/21/2024] [Indexed: 07/12/2024] Open
Abstract
Background Recurrence and metastasis are the major obstacles affecting the therapeutic efficacy and clinical outcomes for patients with esophageal carcinoma (ESCA). Secreted phosphoprotein 1 (SPP1) is considered as a hub gene in ESCA and is negatively associated with disease-free survival (DFS) in ESCA. However, the exact roles and underlying mechanisms remain elusive. This study aims to examine the roles of SPP1 on ESCA, and elucidate the potential mechanisms. Methods Bioinformatics were used to analyze the expression of SPP1 in ESCA tissues, and its relations with clinicopathological characteristics and clinical prognosis in patients with ESCA based on The Cancer Genome Atlas (TCGA) dataset. Loss-of-function was conducted to examine the roles of SPP1 on malignant behaviors of ESCA cells by cell counting kit-8 (CCK8), plate clone, wound healing, and transwell assays. Gene set enrichment analysis (GSEA) was conducted to screen the pathways associated with SPP1 in ESCA. Then, the enriched pathway and the underlying mechanism were elucidated by western blotting, cell adhesion, and cell spreading assays. Lastly, Y15 [a specific inhibitor of focal adhesion kinase (FAK)] was used to examine its potential to inhibit tumor growth in ESCA cells. Results SPP1 was upregulated in ESCA tissues compared to the adjacent nontumorous tissues, which was closely associated with clinical stage, lymph node metastasis, histological subtype, and p53 mutation. A high expression of SPP1 indicated a poor clinical prognosis in patients with ESCA. The knockdown of SPP1 inhibited cell proliferative, migratory, and invasive capacities in ESCA cells. GSEA indicated that the focal adhesion pathway was closely related with SPP1 in ESCA. Further studies confirmed that the knockdown of SPP1 suppressed cell adhesion ability and reduced the expression of p-FAK and p-Erk in ESCA cells. In addition, Y15 inhibited FAK autophosphorylation and dramatically inhibited cell proliferation, migration, and invasion in ESCA cells. Conclusions SPP1 promotes tumor progression in ESCA by activating FAK/Erk pathway, and FAK is a potential therapeutic target to overcome tumor recurrence and metastasis of ESCA.
Collapse
Affiliation(s)
- Jianjun Sun
- Department of Thoracic Surgery, Naval Specialized Medical Center Affiliated to Naval Medical University, Shanghai, China
| | - Mingming Tang
- Department of Thoracic Surgery, Naval Specialized Medical Center Affiliated to Naval Medical University, Shanghai, China
| | - Zhigang Cai
- Department of Thoracic Surgery, Naval Specialized Medical Center Affiliated to Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Che XQ, Zhan SK, Song JJ, Deng YL, Wei-Liu, Peng-Huang, Jing-Zhang, Sun ZF, Che ZQ, Liu J. Altered immune pathways in patients of temporal lobe epilepsy with and without hippocampal sclerosis. Sci Rep 2024; 14:13661. [PMID: 38871732 PMCID: PMC11176392 DOI: 10.1038/s41598-024-63541-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
Over the past decades, the immune responses have been suspected of participating in the mechanisms for epilepsy. To assess the immune related pathway in temporal lobe epilepsy (TLE), we explored the altered immune pathways in TLE patients with and without hippocampal sclerosis (HS). We analyzed RNA-seq data from 3 TLE-HS and 3 TLE-nonHS patients, including identification of differentially expressed RNA, function pathway enrichment, the protein-protein interaction network and construction of ceRNA regulatory network. We illustrated the immune related landscape of molecules and pathways on human TLE-HS. Also, we identified several differential immune related genes like HSP90AA1 and SOD1 in TLE-HS patients. Further ceRNA regulatory network analysis found SOX2-OT connected to miR-671-5p and upregulated the target gene SPP1 in TLE-HS patients. Also, we identified both SOX2-OT and SPP1 were significantly upregulated in five different databases including TLE-HS patients and animal models. Our findings established the first immune related genes and possible regulatory pathways in TLE-HS patients and animal models, which provided a novel insight into disease pathogenesis in both patients and animal models. The immune related SOX2-OT/miR-671-5p/SPP1 axis may be the potential therapeutic target for TLE-HS.
Collapse
Affiliation(s)
- Xiang-Qian Che
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shi-Kun Zhan
- Department of Neurosurgery, Centre for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao-Jiao Song
- Department of Teaching Office, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Lei Deng
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Liu
- Department of Neurosurgery, Centre for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng-Huang
- Department of Neurosurgery, Centre for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Zhang
- Department of Neurosurgery, Centre for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhan-Fang Sun
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zai-Qian Che
- Department of Emergency, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Jun Liu
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Koroknai V, Szász I, Jámbor K, Balázs M. Expression pattern of osteopontin isoforms in malignant melanoma cell lines. Clin Transl Sci 2024; 17:e13694. [PMID: 38058256 PMCID: PMC10772848 DOI: 10.1111/cts.13694] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023] Open
Abstract
Osteopontin (OPN) is a secreted integrin-binding protein that plays a role in inflammation, cellular viability, cell adhesion and migration, cancer development, and diabetes through different mechanisms. The splice variants of OPN can play essential roles in cancer development, progression, and metastasis formation; however, limited data are available about the role of OPN isoforms in human malignant melanoma. Our goal was to define the gene expression patterns of five OPN variants (OPN4, OPN5, OPNa, OPNb, and OPNc), integrin, and CD44 receptor genes in primary and metastatic melanoma-originated cell lines (n = 19), and to explore the association of the expression patterns with clinicopathological parameters. We evaluated the invasive property of the cell lines and investigated the potential association between the invasion and gene expression of OPN isoforms. We found a significant rise in the expression of OPNc in the invasive cell lines compared to the noninvasive cells and detected significantly higher expression of the OPN splice variants in melanoma cell lines originating from more advanced stages tumors than cell lines originating from early-stage melanomas. The correlation analysis revealed that all five OPN variants positively correlated with ITGB3 and ITGA9, whereas OPN5 positively correlated with ITGB1, ITGAV, ITGA6, and CD44. OPN can activate extracellular signal-regulated kinase signaling through binding to α9β1 integrin, promoting melanoma tumor cell migration. It is possible that such associations between OPN splice variants and integrin receptors may play a role in melanoma progression. In conclusion, our findings suggest that high expression of OPNc correlates with the invasive behavior of melanoma cells.
Collapse
Affiliation(s)
- Viktoria Koroknai
- Department of Public Health and Epidemiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- HUN‐REN‐DE Public Health Research GroupUniversity of DebrecenDebrecenHungary
| | - István Szász
- Department of Public Health and Epidemiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- HUN‐REN‐DE Public Health Research GroupUniversity of DebrecenDebrecenHungary
| | - Krisztina Jámbor
- Department of Public Health and Epidemiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
| | - Margit Balázs
- Department of Public Health and Epidemiology, Faculty of MedicineUniversity of DebrecenDebrecenHungary
- HUN‐REN‐DE Public Health Research GroupUniversity of DebrecenDebrecenHungary
| |
Collapse
|
6
|
Lukianova N, Zadvornyi Т, Borikun Т, Mushii О, Pavlova А, Tymoshenko А, Stakhovskyi Е, Vitruk I, Сhekhun V. SIGNIFICANCE OF OSTEOPONTIN FOR PREDICTING AGGRESSIVENESS OF PROSTATE CANCER. Exp Oncol 2023; 45:312-321. [PMID: 38186024 DOI: 10.15407/exp-oncology.2023.03.312] [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: 12/27/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Effective prediction of the course of prostate cancer (PCa) and the stratification of treatment tactics largely depend on the use of prognostic markers that reflect the molecular and biological features of tumors. In view of the important role of matricellular proteins in the modulation of the growing tumor and metastasis of the hormone-dependent neoplasms, the aim of the work was to study the expression of osteopontin (OPN) at the protein and mRNA levels in the PCa tissue in order to assess the significance of this protein for predicting the aggressiveness of PCa. MATERIALS AND METHODS The work is based on the analysis of the results of the examination and treatment of 83 patients with PCa of stages II-IV. The study of OPN expression at the level of mRNA and protein in the PCa tissue was carried out using methods of the real time polymerase chain reaction and immunohistochemistry, respectively. RESULTS The OPN expression in the PCa tissue was 1.6 times (p < 0.05) higher in patients with regional lymph node metastases compared to patients without metastases. In patients with a Gleason score of < 7, the OPN expression in the tumor tissue was 1.4 times lower (p < 0.05) than in patients with poorly differentiated PCa. In patients with a high risk of tumor progression, the OPN expression level was 1.4 and 2.1 times higher (p < 0.05) compared to patients with a moderate and low risk of PCa progression. The patients with a high OPN expression level in the PCa tissue had significantly decreased 2-year recurrence-free survival rate (by 25%). CONCLUSIONS The obtained results indicate the expediency of using OPN expression indicators in the tumor tissue to predict the PCa aggressiveness and assess the risk of its recurrence.
Collapse
Affiliation(s)
- N Lukianova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Т Zadvornyi
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Т Borikun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - О Mushii
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - А Pavlova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - А Tymoshenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Е Stakhovskyi
- The State Non-Profit Enterprise "National Cancer Institute" of the Ministry of Health of Ukraine, Kyiv, Ukraine
| | - I Vitruk
- The State Non-Profit Enterprise "National Cancer Institute" of the Ministry of Health of Ukraine, Kyiv, Ukraine
| | - V Сhekhun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
7
|
Song Y, Wang ZY, Luo J, Han WC, Wang XY, Yin C, Zhao WN, Hu SW, Zhang Q, Li YQ, Cao JL. CWC22-Mediated Alternative Splicing of Spp1 Regulates Nociception in Inflammatory Pain. Neuroscience 2023; 535:50-62. [PMID: 37838283 DOI: 10.1016/j.neuroscience.2023.10.006] [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: 06/06/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/16/2023]
Abstract
Increasing evidence suggests that alternative splicing plays a critical role in pain, but its underlying mechanism remains elusive. Herein, we employed complete Freund's adjuvant (CFA) to induce inflammatory pain in mice. A combination of genomics research techniques, lentivirus-based genetic manipulations, behavioral tests, and molecular biological technologies confirmed that splicing factor Cwc22 mRNA and CWC22 protein were elevated in the spinal dorsal horn at 3 days after CFA injection. Knockdown of spinal CWC22 by lentivirus transfection (lenti-shCwc22) reversed CFA-induced thermal hyperalgesia and mechanical allodynia, whereas upregulation of spinal CWC22 (lenti-Cwc22) in naïve mice precipitated pain. Comprehensive transcriptome and genome analysis identified the secreted phosphoprotein 1 (Spp1) as a potential gene of CWC22-mediated alternative splicing, however, only Spp1 splicing variant 4 (Spp1 V4) was involved in thermal and mechanical nociceptive regulation. In conclusion, our findings demonstrate that spinal CWC22 regulates Spp1 V4 to participate in CFA-induced inflammatory pain. Blocking CWC22 or CWC22-mediated alternative splicing may provide a novel therapeutic target for the treatment of persistent inflammatory pain.
Collapse
Affiliation(s)
- Yu Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, China
| | - Zhi-Yong Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Department of Anesthesiology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210008, China
| | - Jun Luo
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Department of Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221002, China
| | - Wen-Can Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Xiao-Yi Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Department of Anesthesiology, Gulou Hospital Affiliated to Medical College of Nanjing University, Nanjing, Jiangsu Province 210008, China
| | - Cui Yin
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Wei-Nan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Qi Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Yan-Qiang Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China.
| |
Collapse
|
8
|
Silver SV, Popovics P. The Multifaceted Role of Osteopontin in Prostate Pathologies. Biomedicines 2023; 11:2895. [PMID: 38001899 PMCID: PMC10669591 DOI: 10.3390/biomedicines11112895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
The prostate gland, located beneath the bladder and surrounding the proximal urethra in men, plays a vital role in reproductive physiology and sexual health. Despite its importance, the prostate is vulnerable to various pathologies, including prostatitis, benign prostatic hyperplasia (BPH) and prostate cancer (PCa). Osteopontin (OPN), a versatile protein involved in wound healing, inflammatory responses, and fibrotic diseases, has been implicated in all three prostate conditions. The role of OPN in prostatic pathophysiology, affecting both benign and malignant prostate conditions, is significant. Current evidence strongly suggests that OPN is expressed at a higher level in prostate cancer and promotes tumor progression and aggressiveness. Conversely, OPN is primarily secreted by macrophages and foam cells in benign prostate conditions and provokes inflammation and fibrosis. This review discusses the accumulating evidence on the role of OPN in prostatic diseases, cellular sources, and potential roles while also highlighting areas for future investigations.
Collapse
Affiliation(s)
- Samara V. Silver
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Petra Popovics
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| |
Collapse
|
9
|
An Y, Fnu G, Xie C, Weber GF. Meta-analysis of Osteopontin splice variants in cancer. BMC Cancer 2023; 23:373. [PMID: 37095438 PMCID: PMC10124019 DOI: 10.1186/s12885-023-10854-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 04/14/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND The cytokine Osteopontin is a mediator of tumor progression and cancer metastasis. In 2006, we reported that (in addition to the full-length form -a) splice variants of Osteopontin (forms -b and -c) are produced selectively by transformed cells. Through June 2021, 36 PubMed-indexed journal articles have studied Osteopontin splice variants in various cancer patients. METHODS Applying a categorical approach previously developed by us, here we conduct a meta-analysis of the pertinent literature. We supplement this with evaluation of the relevant entries in the TSVdb database, which focusses on splice variant expression, thus including the additional variants -4 and -5. The analysis covers 5886 patients across 15 tumors from the literature and 10,446 patients across 33 tumors from TSVdb. RESULTS The database yields positive results more frequently than the categorical meta-analysis. The two sources are in agreement on the elevation of OPN-a, OPN-b, and OPN-c in lung cancer and the elevation of OPN-c in breast cancer as compared to healthy tissue. Specific splice variants are associated with grade, stage, or patient survival pertaining to various cancers. CONCLUSIONS There are cases of persisting discrepancies, which require further investigation to clarify the Osteopontin splice variant utilization, so that their diagnostic, prognostic and potentially predictive potential can be brought to fruition.
Collapse
Affiliation(s)
- Yu An
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Gulimirerouzi Fnu
- James L. Winkle College of Pharmacy, College of Pharmacy, University of Cincinnati Academic Health Center, 231 Albert Sabin Way, Cincinnati, OH, USA
| | - Changchun Xie
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Georg F Weber
- James L. Winkle College of Pharmacy, College of Pharmacy, University of Cincinnati Academic Health Center, 231 Albert Sabin Way, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Understanding of molecular basis of histological graded horn cancer by transcriptome profiling. Gene 2023; 857:147196. [PMID: 36641075 DOI: 10.1016/j.gene.2023.147196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/21/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Horn cancer is most devastating and prominent cancer in Indian zebu cattle that affects socio-economic condition of small-scale farmers who depends on their cattle for farm work. Development in the field for genomics through next generation sequencing and bioinformatics advancement have helped to identify genes which have a role in horn cancer development. Histopathological examination of cancerous tissues of horn revealed myxomatous changes, well, moderate and poorly differentiated squamous cell carcinoma. Differential gene expression analysis showed 40, 11, 66 and 29 upregulated genes and 10, 14, 08 and 07 down-regulated genes in myxomatous, well, moderate and poorly differentiated squamous cell carcinoma as compared to normal. Significant differentially expressed genes are related to cell development, cell proliferation, cell-cell communication, cell signaling and angiogenesis which are linked to Akt pathway, mTOR pathway and Wnt pathway. Activity of these genes and related pathways have already been established about their role in development of cancer. Among the candidate genes; keratin family, keratin family related gene, chemokine signaling and cytokines signaling associated genes could be a prominent target for the development of stage specific prognosis marker after further detailed study at large sample population level. CSTA, PTN, SPP1 genes have upregulation in all stages of cancer and they have enrolled as biomarkers for horn cancer.
Collapse
|
11
|
The Intracellular and Secreted Sides of Osteopontin and Their Putative Physiopathological Roles. Int J Mol Sci 2023; 24:ijms24032942. [PMID: 36769264 PMCID: PMC9917417 DOI: 10.3390/ijms24032942] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Classically, osteopontin (OPN) has been described as a secreted glycophosprotein. Indeed, most data concerning its physiological and pathological roles are mainly related to the secreted OPN (sOPN). However, there are several instances in which intracellular OPN (iOPN) has been described, presenting some specific roles in distinct experimental models, such as in the immune system, cancer cells, and neurological disorders. We herein aimed to highlight and discuss some of these secreted and intracellular roles of OPN and their putative clinical and biological impacts. Moreover, by consolidating data from the OPN protein database, we also analyzed the occurrence of signal peptide (SP) sequences and putative subcellular localization, especially concerning currently known OPN splicing variants (OPN-SV). Comprehending the roles of OPN in its distinct cellular and tissue environments may provide data regarding the additional applications of this protein as biomarkers and targets for therapeutic purposes, besides further describing its pleiotropic roles.
Collapse
|
12
|
Yimingjiang M, Aini A, Tuergan T, Zhang W. Differential Gene Expression Profiling in Alveolar Echinococcosis Identifies Potential Biomarkers Associated With Angiogenesis. Open Forum Infect Dis 2023; 10:ofad031. [PMID: 36817746 PMCID: PMC9927572 DOI: 10.1093/ofid/ofad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Background Alveolar echinococcosis (AE) is a worldwide zoonosis caused by Echinococcus multilocularis. Alveolar echinococcosis is a severe chronic parasitic disease that exhibits a tumor-like growth, with the potential for invasion and distant metastasis; however, the molecular mechanism underlying this condition remains unclear. Methods Transcriptome analyses were performed to detect differentially expressed genes (DEGs) in samples from patients with AE with invasion and distant metastasis. The results were further verified by immunohistochemistry. Results A total of 1796 DEGs were identified, including 1742 upregulated and 54 downregulated DEGs. A subsequent functional analysis showed that the significant DEGs were involved in the angiogenesis process. Immunohistochemical analysis confirmed the reliability of the transcriptomic data. Conclusions These results suggest that angiogenesis is a possible mechanism underlying the tumor-like biological behavior observed during E multilocularis infection. Genes related to this process may play important roles in AE invasion and distant metastasis.
Collapse
Affiliation(s)
- Maiweilidan Yimingjiang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Abudusalamu Aini
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Talaiti Tuergan
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Wei Zhang
- Correspondence: Dr. Wei Zhang, Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 137 Liyushan Southern Road, Urumqi, Xinjiang 830054, China ( )
| |
Collapse
|
13
|
Osteopontin Splicing Isoforms Contribute to Endometriotic Proliferation, Migration, and Epithelial-Mesenchymal Transition in Endometrial Epithelial Cells. Int J Mol Sci 2022; 23:ijms232315328. [PMID: 36499654 PMCID: PMC9738877 DOI: 10.3390/ijms232315328] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN) isoforms, including OPNb and OPNc, promote malignancy and may contribute to the pathogenesis of endometriosis, a benign disorder with multiple characteristics resembling malignant tumors. In our experiments, OPNb and OPNc were significantly overexpressed in both endometriosis and adenomyosis compared to the normal endometrium. Upregulation of CD44v and the epithelial-mesenchymal transition (EMT) process was also present in endometriotic lesions. Overexpression of OPNb and OPNc splicing variants in endometriotic cells evoked morphological changes, actin remodeling, cell proliferation, cell migration, and EMT through binding OPN ligand receptors CD44 and αvβ3, subsequently activating the PI3K and NF-ĸB pathways. We elucidated the causal role of OPN splice variants in regulating endometriotic cell growth, which may promote the development of OPN-targeted therapies for patients suffering from endometriotic disorders.
Collapse
|
14
|
Yi X, Luo L, Zhu Y, Deng H, Liao H, Shen Y, Zheng Y. SPP1 facilitates cell migration and invasion by targeting COL11A1 in lung adenocarcinoma. Cancer Cell Int 2022; 22:324. [PMID: 36266702 PMCID: PMC9583566 DOI: 10.1186/s12935-022-02749-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background Secreted phosphoprotein 1 (SPP1), an extracellular secreted glycol phosphoprotein, is closely related to tumor biologies, such as proliferation, migration, and invasion. However, the role and biological function of SPP1 in lung adenocarcinoma (LUAD) was still ambiguous. Methods SPP1 expression in LUAD tissues and its associations with clinical features and prognosis was investigated using meta-analysis, immunohistochemistry (IHC) staining methods, and quantitative real-time polymerase chain reaction (qRT-PCR). Moreover, the potential mechanism related to SPP1 was identified by using the Gene Set Enrichment Analysis (GSEA) method. A series of function assays were conducted to determine the biological role of SPP1 in LUAD cell migration and invasion in vitro and vivo. The co-expressed genes of SPP1 were obtained and verified by western blot assays. The influence of SPP1 on Collagen type XI alpha 1 (COL11A1) expression and epithelial-mesenchymal transition (EMT) markers was analyzed using western blot assays. Results The expression of SPP1 in LUAD tissues and cells was significantly higher than that in normal tissues and cells. And positively associations of SPP1 expression with TNM stage, lymph node metastasis, and invasion depth were observed. Patients with high SPP1 expression had unfavorable survival. The multivariable Cox regression analysis revealed that SPP1 expression was an independent prognostic factor of LUAD patients. Furthermore, downregulation of SPP1 could inhibit cell migration and invasion both in vitro and vivo, reduce the expression of epithelial marker (E-cadherin), and increase the expression of mesenchymal markers (N-cadherin and vimentin). Using bioinformatics and western blot assays, we confirmed that COL11A1 acted as the downstream of SPP1, and SPP1 knockdown could significantly downregulate the COL11A1 expression. Importantly, suppression of cell migration and invasion and the expression changes of EMT markers induced by SPP1 downregulation could be reversed by COL11A1 overexpression. Conclusions SPP1 facilitates cell migration and invasion by upregulating COL11A1 expression and that acts as a potential biomarker of metastasis and prognosis for LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02749-x.
Collapse
Affiliation(s)
- Xuan Yi
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.,Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Linlin Luo
- Second Department of Respiratory Disease, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330000, China
| | - Yanzhen Zhu
- Department of Anesthesia and Perioperative Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Hong Deng
- Pharmacy Department, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, 332000, China
| | - Huitian Liao
- Financial Department, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Yang Shen
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.,Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Yan Zheng
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China. .,Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
| |
Collapse
|
15
|
Kamalabadi-Farahani M, Atashi A, Jabbarpour Z, Aghayan SS. Expression of osteopontin-5 splice variant in the mouse primary and metastatic breast cancer cells. BMC Res Notes 2022; 15:286. [PMID: 36064446 PMCID: PMC9446537 DOI: 10.1186/s13104-022-06179-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Osteopontin (OPN) is a well-known glycoprotein involved in numerous pathobiological processes, including cancer. Despite having five splice variants for osteopontin in mice, the main focus of most studies has been on total OPN (tOPN). There are some studies on other splice variants, but the expression of osteopontin-5 (OPN5) has not been addressed in mouse cancer cells. Therefore, this study sought to evaluate OPN5 expression in mouse breast cancer cells. RESULTS The expression of OPN5 in primary and metastatic breast cancer cells of mice was confirmed in our study. These findings provided important insights regarding the OPN alternative splicing in mice for the first time. It is concluded that, like other OPN-SVs, OPN5 probably plays an essential role in tumor progression, which requires further investigation in different tumor models.
Collapse
Affiliation(s)
| | - Amir Atashi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Zahra Jabbarpour
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Sajjad Aghayan
- Gene Therapy Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Jámbor K, Koroknai V, Kiss T, Szász I, Pikó P, Balázs M. Gene Expression Patterns of Osteopontin Isoforms and Integrins in Malignant Melanoma. PATHOLOGY AND ONCOLOGY RESEARCH 2022; 28:1610608. [PMID: 36091936 PMCID: PMC9448871 DOI: 10.3389/pore.2022.1610608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
Abstract
Osteopontin (OPN) is a multifunctional glycoprotein that physiologically interacts with different types of integrins. It is considered to be a possible prognostic biomarker in certain tumor types; however, various splicing isoforms exist, which have not been investigated in melanoma. We aimed to define the relative expression pattern of five OPN isoforms and clarify the prognostic significance of the splice variants in melanoma. We also aimed to investigate the expression pattern of eight integrins in the same tumors. Gene expression analyses revealed that the relative expression of OPNa, OPNb, and OPNc is significantly higher in metastatic tumors compared to primary lesions (p < 0.01), whereas the expression of OPN4 and OPN5 was low in both. The more aggressive nodular melanomas had higher expression levels compared to the superficial spreading subtype (p ≤ 0.05). The relative expression of the eight tested integrins was low, with only the expression of ITGB3 being detectable in nodular melanoma (Medianlog2 = 1.274). A positive correlation was found between Breslow thickness and the expression of OPNc variant, whereby thicker tumors (>4 mm) had significantly higher expression (p ≤ 0.05). The Breslow thickness was negatively correlated with the expression of OPN4, and similarly with ITGA2. OPNc also exhibited significant positive correlation with the presence of metastasis. Our data show that high expression of OPNa, OPNb, and especially OPNc and low expression of OPN4 and ITGA2 are associated with an advanced stage of tumor progression and poor prognosis in melanoma.
Collapse
Affiliation(s)
- Krisztina Jámbor
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktória Koroknai
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Tímea Kiss
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Szász
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Péter Pikó
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Margit Balázs
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
- *Correspondence: Margit Balázs,
| |
Collapse
|
17
|
Wang M, Sun X, Xin H, Wen Z, Cheng Y. SPP1 promotes radiation resistance through JAK2/STAT3 pathway in esophageal carcinoma. Cancer Med 2022; 11:4526-4543. [PMID: 35593388 PMCID: PMC9741975 DOI: 10.1002/cam4.4840] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Therapeutic resistance to radiotherapy is one of the major obstacles in clinical practice that significantly affect the therapeutic efficiency and prognosis of human esophageal carcinoma (ESCA). Thus, it is critical to understand the molecular mechanisms of radiation resistance in ESCA. Secreted phosphoprotein 1 (SPP1) plays an essential role in various human cancers, but its role in radiation resistance remains unclear. METHOD Cell culture and transfection; Cell Counting Kit-8 (CCK-8) assays; EdU incorporation assays; Patient sample collection and medical records review; Transwell assays; Colony formation assays; Wound healing assays; Western blot; Immunofluorescence; Immunohistochemistry; Irradiation; Flow cytometry; Animal studies; Human Apoptosis Array Kit; Bioinformatics. RESULT In the current study, we reported the novel phenomenon that radiation-treated human ESCA cells upregulated SPP1 expression, which in turn contributed to the ESCA resistance to radiotherapy. We also reported the tumor-promoting effect of SPP1 in ESCA systematically and comprehensively. Furthermore, subsequent studies by knocking down or overexpressing SPP1 in human ESCA cells showed that SPP1 could facilitate the repair of DNA damage and the survival of tumor cells post-radiation in ESCA, which might contribute to the development of radiation resistance during the radiotherapy process. More detailed investigations on the downstream molecular pathway suggested that radiation could increase the phosphorylation level of JAK2 and STAT3 by increasing SPP1 expression. Further in vivo validation using a mouse ESCA xenograft model showed that SPP1 overexpression significantly increased tumor volume while either SPP1 knockdown or pharmacological inhibition of the JAK2-STAT3 pathway reduced tumor volume in a synergistic manner with radiotherapy. CONCLUSION Collectively, these findings suggested that the SPP1/JAK2/STAT3 axis is a critical player in ESCA progression and radiation resistance, which is a potential therapeutic target for combined therapy with the standard radiotherapy regimen to improve curative effect and increase patients' survival with ESCA.
Collapse
Affiliation(s)
- Meijie Wang
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Xiaozheng Sun
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Huixian Xin
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Zhihua Wen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| |
Collapse
|
18
|
Shao Z, Bi S. Endocrine regulation and metabolic mechanisms of osteopontin in the development and progression of osteosarcoma, metastasis and prognosis. Front Endocrinol (Lausanne) 2022; 13:1100063. [PMID: 36714568 PMCID: PMC9880040 DOI: 10.3389/fendo.2022.1100063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Osteosarcoma is the most common type of malignant bone tumor, occurring in adolescents and patients over 60. It has a bimodal onset and a poor prognosis, and its development has not yet been fully explained. Osteopontin (OPN) is a high protein consisting of 314 amino acid residues with a negative charge and is involved in many biological activities. OPN is not only an essential part of the regulation of the nervous system and endocrine metabolism of skeletal cells. Still, it is also involved in several other important biological activities, such as the division, transformation, and proliferation of skeletal cells and their associated cells, such as bone tumor cells, including bone marrow mesenchymal stem cells, hematopoietic stem cells, osteoblasts, and osteoclasts. Osteoblasts and osteocytes. Recent studies have shown a strong correlation between OPN and the development and progression of many skeletal diseases, such as osteosarcoma and rheumatoid arthritis. This review aims to understand the mechanisms and advances in the role of OPN as a factor in the development, progression, metastasis, and prognosis of osteosarcoma in an attempt to provide a comprehensive summary of the mechanisms by which OPN regulates osteosarcoma progression and in the hope of contributing to the advancement of osteosarcoma research and clinical treatment.
Collapse
|
19
|
Panda A, Bhanot G, Ganesan S, Bajpai M. Gene Expression in Barrett's Esophagus Cell Lines Resemble Esophageal Squamous Cell Carcinoma Instead of Esophageal Adenocarcinoma. Cancers (Basel) 2021; 13:5971. [PMID: 34885081 PMCID: PMC8656995 DOI: 10.3390/cancers13235971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is strongly associated with Barrett's esophagus (BE), a pre-malignant condition resulting from gastric reflux. Esophageal squamous cell carcinoma (ESCC), the other major subtype of esophageal cancer, shows strong association with smoking and alcohol intake and no association with gastric reflux. In this study, we constructed and validated gene expression signatures of EAC vs. ESCC tumors using publicly available datasets, and subsequently assessed the enrichment levels of these signatures in commonly used EAC and ESCC cell lines, normal esophageal tissues and normal esophageal cell lines, and primary BE tissues and BE cell lines. We found that unlike ESCC cell lines which were quite similar to primary ESCC tumors, EAC cell lines were considerably different from primary EAC tumors but still more similar to EAC tumors than ESCC tumors, as the genes up in EAC vs. ESCC (EAChi) had considerably lower expression in EAC cell lines than EAC tumors. However, more surprisingly, unlike various normal cell lines (EPC2, Het-1A) which were very similar to various tissues from normal esophagus, BE cell lines (BAR-T, CP-A) were extremely different from primary BE tissues, as BE cell lines had substantially lower levels of EAChi and substantially higher levels of ESCChi gene expression. This ESCC-like profile of the BAR-T remained unaltered even after prolonged exposure to an acidic bile mixture in vitro resulting in malignant transformation of this cell line. However, primary BE tissues had EAC-like gene expression profiles as expected. Only one EAC case from the Cancer Genome Atlas resembled BE cell lines, and while it had the clinical profile and some mutational features of EAC, it had some mutational features, the copy number alteration profile, and the gene expression profile of ESCC instead. These incomprehensible changes in gene expression patterns may result in ambiguous changes in the phenotype and warrants careful evaluation to inform selection of appropriate in vitro tools for future studies on esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Anshuman Panda
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (A.P.); (G.B.); (S.G.)
| | - Gyan Bhanot
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
- Department of Physics and Astronomy, Rutgers University, Piscataway, NJ 08854, USA
| | - Shridar Ganesan
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; (A.P.); (G.B.); (S.G.)
- Center for Systems and Computational Biology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine—Medical Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Manisha Bajpai
- Department of Medicine—Gastroenterology and Hepatology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Cancer Pharmacology Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
| |
Collapse
|
20
|
Hoefnagel SJM, Boonstra JJ, Russchen MJAM, Krishnadath KK. Towards Personalized Treatment Strategies for Esophageal Adenocarcinoma; A Review on the Molecular Characterization of Esophageal Adenocarcinoma and Current Research Efforts on Individualized Curative Treatment Regimens. Cancers (Basel) 2021; 13:4881. [PMID: 34638363 PMCID: PMC8508226 DOI: 10.3390/cancers13194881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/18/2022] Open
Abstract
Esophageal cancers confer a major health challenge and are highly aggressive malignancies with poor prognosis. Esophageal adenocarcinoma (EAC) is one of the two major histopathological subtypes of esophageal cancer. Despite advances in treatment modalities, the prognosis of patients with EAC remains poor, with a 5-year survival rate that rarely exceeds 30% in patients treated with curative intent. Chemoradiotherapy followed by resection is the treatment of choice for EAC patients, which are deemed to be curable. Current patient stratification and treatments are based on outcomes from clinical trials. Unfortunately, the molecular heterogeneity of EAC which determines the chemo- and radiosensitivity of these cancers are not taken into account. A more personalized approach in the treatment of EAC could improve patient outcomes. This review aims at summarizing literature on translational and clinical research in the field of EAC which could be of importance to develop personalized approaches. As suggested by the TCGA, expression data features molecular classifications by different platforms, including miRNA, genomic mutations and reverse-phase protein arrays. Here, we summarize literature on transcriptomic, data-driven approaches to identify distinct subtypes of EAC associated with molecular features. These novel classifications may determine the responsiveness to chemo(radio)therapy and help to identify novel molecular targets within cell signaling pathways. Moreover, we discuss the current clinical research efforts on tailored treatment regimens for patients with EAC taking into account the heterogeneous response to chemoradiotherapy. We summarize the evidence regarding active surveillance instead of immediate surgical resection after application of neoadjuvant chemo(radio)therapy in EAC. We consider that in future patients with complete response to chemo(radio)therapy, predicted by (transcriptomic) biomarkers, might benefit most from this approach. Finally, challenges to overcome for current findings to be implemented in clinical practice and move the field forward are being discussed.
Collapse
Affiliation(s)
- Sanne J. M. Hoefnagel
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Jurjen J. Boonstra
- Department of Gastroenterology and Hepatology, Leiden UMC, 2333 ZA Leiden, The Netherlands;
| | | | - Kausilia K. Krishnadath
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Paediatrics, University of Antwerp, 2000 Antwerpen, Belgium
| |
Collapse
|
21
|
Distinctive Prognostic Value and Cellular Functions of Osteopontin Splice Variants in Human Gastric Cancer. Cells 2021; 10:cells10071820. [PMID: 34359989 PMCID: PMC8305399 DOI: 10.3390/cells10071820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 01/25/2023] Open
Abstract
Background: Osteopontin (OPN) splice variants are identified as predictors of tumour progression and therapeutic resistance in certain types of solid tumours. However, their roles in gastric cancer (GC) remain poorly characterized. The current study sought to assess the prognostic value of the three OPN splice variants (namely OPN-a, OPN-b, and OPN-c) in gastric cancer and their potential functions within gastric cancer cells. Methods: RNA extraction and reverse transcription were performed using our clinical cohort of gastric carcinomas and matched normal tissues (n = 324 matched pairs). Transcript levels were determined using real-time quantitative PCR. Three OPN splice variants overexpressed cell lines were created from the gastric cancer cell line HGC-27. Subsequently, biological functions, including cell growth, adhesion, migration, and invasion, were studied. The potential effects of OPN isoforms on cisplatin and 5-Fu were evaluated by detecting cellular reactive oxygen species (ROS) levels in the HGC-27-derived cell lines. Results: Compared with normal tissues, the expression levels of three splice variants were all elevated in gastric cancer tissues in an order of OPN-a > OPN-b > OPN-c. The OPN-a level significantly increased with increasing TNM staging and worse clinical outcome. There appeared to be a downregulation for OPN-c in increasing lymph node status (p < 0.05), increasing TNM staging, and poor differentiation. High levels of OPN-a and OPN-b were correlated with short overall survival and disease-free survival of gastric cancer patients. However, the low expression of OPN-c was significantly associated with a poor prognosis. Functional analyses further showed that ectopic expression of OPN-c suppressed in vitro proliferation, adhesiveness, migration, and invasion properties of HGC-27 cells, while the opposite role was seen for OPN-a. Cellular ROS detection indicated that OPN-a and OPN-c significantly promoted ROS production after treatment with 5-Fu comparing to OPN-vector, while only OPN-a markedly induced ROS production after treatment with cisplatin. Conclusion: Our results suggest that OPN splice variants have distinguished potential to predict the prognosis of gastric cancer. Three OPN variants exert distinctive functions in gastric cancer cells. Focusing on specific OPN isoforms could be a novel direction for developing diagnostic and therapeutic approaches in gastric cancer.
Collapse
|
22
|
Chou CF, Huang CC, Bin Dabil N, Chang PL. Assessing SPP1/Osteopontin (OPN) Splice Variants and Their Association to Nonmelanoma Skin Cancer by Absolute Quantification: Identification of OPN-5 Subvariants and Their Protein Coding Potential. Cancer Invest 2021; 39:559-570. [PMID: 34043476 DOI: 10.1080/07357907.2021.1933015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The study evaluated whether SPP1/osteopontin (OPN) splice variants are differentially expressed in nonmelanoma skin cancer compared to normal skin. The absolute number of mRNA molecules of OPN-a predominated in normal skin and nonmelanoma skin cancer compared to OPN-b, OPN-c, and OPN-5. However, mRNAs of OPN-a, OPN-b, and OPN-c were expressed in higher levels in cutaneous squamous cell carcinomas (cSCCs) and basal cell carcinomas relative to normal skin. Additionally, OPN-5 expression was higher than OPN-b and OPN-c, and OPN-c, in normal skin and nonmelanoma skin cancer, respectively. Furthermore, we identified four OPN-5 splice variants, which were cloned and analyzed for protein expression.
Collapse
Affiliation(s)
- Chu-Fang Chou
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Conway C Huang
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Noura Bin Dabil
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Pi-Ling Chang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
23
|
Identification of crucial long non-coding RNAs and mRNAs along with related regulatory networks through microarray analysis in esophageal carcinoma. Funct Integr Genomics 2021; 21:377-391. [PMID: 33864185 DOI: 10.1007/s10142-021-00784-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/01/2023]
Abstract
Esophageal carcinoma (EC) is a tremendous threat to human health and life worldwide. Long non-coding RNAs (lncRNAs) have been identified as crucial players in carcinomas including EC. An in-depth understanding on regulatory networks of lncRNAs contributes to the better management of EC. In this text, 2052 lncRNAs and 3240 mRNAs were found to be differentially expressed in 5 EC tumor tissues versus adjacent normal tissues by microarray analysis. Moreover, 297 carcinoma-related genes were screened out according to pathway and disease annotation analyses. In addition, 410 potential lncRNA-mRNA cis-regulation pairs and 395 lncRNA-mRNA trans-regulation pairs were screened out. Among these genes, 14 trans-regulated and 19 cis-regulated genes were found to be related with carcinomas. Additionally, 42 possible lncRNA-mRNA trans-regulation pairs and 26 cis-regulation pairs were found to be related with carcinomas. Also, 4 differentially expressed transcription factors in EC and lncRNAs possibly regulated by these transcription factors were screened out. Moreover, plenty of common upregulated or downregulated lncRNAs and mRNAs in EC were identified by comparative analysis for our microarray outcomes and previous high-throughput data. Furthermore, we demonstrated that ENST00000437781.1 knockdown inhibited cell proliferation and facilitated cell apoptosis by downregulating SIX homeobox 4 (SIX4) and ENST00000524987.1 knockdown had no influence on anoctamin 1 calcium activated chloride channel (ANO1) expression in EC cells. In conclusion, we identified some crucial lncRNAs and genes along with potential regulatory networks of lncRNAs/genes, deepening our understanding on pathogenesis of EC.
Collapse
|
24
|
The Roles of Osteopontin in the Pathogenesis of West Nile Encephalitis. Vaccines (Basel) 2020; 8:vaccines8040748. [PMID: 33317005 PMCID: PMC7768535 DOI: 10.3390/vaccines8040748] [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: 10/14/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/18/2022] Open
Abstract
Osteopontin (OPN), a multifunctional protein encoded by the secreted phosphoprotein-1 (Spp-1) gene in humans, plays important roles in a variety of physiological conditions, such as biomineralization, bone remodeling and immune functions. OPN also has significant roles in the pathogenesis of autoimmune, allergy and inflammatory diseases, as well as bacterial, fungal and viral infections. West Nile virus (WNV), a mosquito-transmitted flavivirus, is the leading agent for viral encephalitis in North America. Recent progress has been made in understanding both the biological functions of OPN and the pathogenesis of WNV. In this review article, we have summarized the current understanding of the biology of OPN and its vital roles in the pathogenesis of WNV encephalitis.
Collapse
|
25
|
Silva GR, Mattos DS, Bastos ACF, Viana BPPB, Brum MCM, Ferreira LB, Gimba ERP. Osteopontin-4 and Osteopontin-5 splice variants are expressed in several tumor cell lines. Mol Biol Rep 2020; 47:8339-8345. [PMID: 33006711 DOI: 10.1007/s11033-020-05867-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Among osteopontin splice variants (OPN-SV), the expression profile of osteopontin-4 (OPN4) and osteopontin-5 (OPN5) has not been addressed in distinct cancer types. We herein aimed to investigate their expression in several cancer cell lines, besides comparing it in relation to the three previously described OPN-SV: OPNa, OPNb and OPNc. Total RNA from cancer cell lines, including prostate (PC3 and DU145), ovarian (A2780), breast (MCF-7 and MDA-MB-231), colorectal (Caco-2, HT-29 and HCT-116), thyroid (TT, TPC1 and 8505c) and lung (A549 and NCI-H460) was extracted, followed by cDNA synthesis. OPN-SV transcript analysis by RT-PCR or RT-qPCR were performed using OPN-SV specific oligonucleotides and gapdh and actin transcripts were used as housekeeping controls. OPN4 and OPN5 transcripts displayed co-expression in most tested cell lines. OPN4 was found expressed in similar or higher levels in relation to OPN5. Moreover, in most tested cell lines, OPN4 is also expressed in similar levels to OPNa or OPNb. The expression of OPN5 is also generally variable in relation to the other OPN-SV, but expressed in similar or higher levels in relation to OPNc, depending on each tested cell line. OPN4 and OPN5 seem to be co-expressed in several tumor types and OPN4 is one of the most overexpressed OPN-SV in distinct tumor cell lines. Once both OPN4 and OPN5 are differentially expressed and also evidence tumor-specific expression patterns, we hypothesize that similarly to the other OPN-SV, they also possibly contribute to key aspects of tumor progression, what should be further functionally investigated in distinct tumor models.
Collapse
Affiliation(s)
- Gabriela Ribeiro Silva
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil.,Programa de Pós-Graduação Em Ciências Biomédicas, Fisiologia E Farmacologia, Instituto Biomédico, Av. Prof. Hernani Melo, Niterói, 101, CEP24210-130, Brazil
| | - Daniella Santos Mattos
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil.,Programa de Pós-Graduação Stricto Sensu em Oncologia, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil
| | - Ana Clara Fonseca Bastos
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil.,Programa de Pós-Graduação Stricto Sensu em Oncologia, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil
| | - Bruna Prunes Pena Baroni Viana
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil.,Programa de Pós-Graduação Stricto Sensu em Oncologia, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil
| | - Mariana Concentino Menezes Brum
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil.,Programa de Pós-Graduação Stricto Sensu em Oncologia, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil
| | - Luciana Bueno Ferreira
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil.,Programa de Pós-Graduação Stricto Sensu em Oncologia, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil
| | - Etel Rodrigues Pereira Gimba
- Programa de Oncobiologia Celular e Molecular, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil. .,Programa de Pós-Graduação Stricto Sensu em Oncologia, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil. .,Divisão de Pesquisa Clínica, Instituto Nacional de Câncer, Rua André Cavalcanti, 37, 3° andar, Rio de Janeiro, CEP: 20 231 050, Brazil. .,Departamento de Ciências da Natureza, Instituto de Humanidades E Saúde, Universidade Federal Fluminense, Rua Recife 1-7, Bairro Bela Vista, Rio das Ostras, RJ, CEP 28880-000, Brazil. .,Programa de Pós-Graduação Em Ciências Biomédicas, Fisiologia E Farmacologia, Instituto Biomédico, Av. Prof. Hernani Melo, Niterói, 101, CEP24210-130, Brazil.
| |
Collapse
|
26
|
RNA binding proteins: Linking mechanotransduction and tumor metastasis. Cancer Lett 2020; 496:30-40. [PMID: 33007411 DOI: 10.1016/j.canlet.2020.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/07/2023]
Abstract
Mechanotransduction is the leading cellular process that mammalian cells adopted to receive and respond to various mechanical cues from their local microenvironment. Increasing evidence suggests that mechano-transduction is involved in many physiological and disease conditions, ranging from early embryonic development, organogenesis, to a variety of human diseases including cancer. Mechanotransduction is mediated through several classes of senor proteins on the cell surface, intracellular signaling mediators, and core transcriptional regulation networks. Dissecting the molecular mechanisms regulating mechanotransduction and their association with cancer metastasis has received much attention in recent years. RNA binding proteins (RBPs) are a special group of nucleic acid interacting factors that participate in many important cellular processes. In this review, we would like to summarize recent research progresses in understanding the role of RBPs-mediated regulation in mechanotransduction and cancer metastasis. Those intriguing findings will provide novel insights for the disease and guide the potential development of new therapeutic approaches.
Collapse
|
27
|
Nangraj AS, Selvaraj G, Kaliamurthi S, Kaushik AC, Cho WC, Wei DQ. Integrated PPI- and WGCNA-Retrieval of Hub Gene Signatures Shared Between Barrett's Esophagus and Esophageal Adenocarcinoma. Front Pharmacol 2020; 11:881. [PMID: 32903837 PMCID: PMC7438937 DOI: 10.3389/fphar.2020.00881] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/28/2020] [Indexed: 02/05/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is a deadly cancer with high mortality rate, especially in economically advanced countries, while Barrett's esophagus (BE) is reported to be a precursor that strongly increases the risk of EAC. Due to the complexity of these diseases, their molecular mechanisms have not been revealed clearly. This study aims to explore the gene signatures shared between BE and EAC based on integrated network analysis. We obtained EAC- and BE-associated microarray datasets GSE26886, GSE1420, GSE37200, and GSE37203 from the Gene Expression Omnibus and ArrayExpress using systematic meta-analysis. These data were accompanied by clinical data and RNAseq data from The Cancer Genome Atlas (TCGA). Weighted gene co-expression network analysis (WGCNA) and differentially expressed gene (DEG) analysis were conducted to explore the relationship between gene sets and clinical traits as well as to discover the key relationships behind the co-expression modules. A differentially expressed gene-based protein-protein interaction (PPI) complex was used to extract hub genes through Cytoscape plugins. As a result, 403 DEGs were excavated, comprising 236 upregulated and 167 downregulated genes, which are involved in the cell cycle and replication pathways. Forty key genes were identified using modules of MCODE, CytoHubba, and CytoNCA with different algorithms. A dark-gray module with 207 genes was identified which having a high correlation with phenotype (gender) in the WGCNA. Furthermore, five shared hub gene signatures (SHGS), namely, pre-mRNA processing factor 4 (PRPF4), serine and arginine-rich splicing factor 1 (SRSF1), heterogeneous nuclear ribonucleoprotein M (HNRNPM), DExH-Box Helicase 9 (DHX9), and origin recognition complex subunit 2 (ORC2), were identified between BE and EAC. SHGS enrichment denotes that RNA metabolism and splicosomes play a key role in esophageal cancer development and progress. We conclude that the PPI complex and WGCNA co-expression network highlight the importance of phenotypic identifying hub gene signatures for BE and EAC.
Collapse
Affiliation(s)
- Asma Sindhoo Nangraj
- The State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Gurudeeban Selvaraj
- Center of Interdisciplinary Sciences-Computational Life Sciences, Henan University of Technology, Zhengzhou, China
| | - Satyavani Kaliamurthi
- Center of Interdisciplinary Sciences-Computational Life Sciences, Henan University of Technology, Zhengzhou, China
| | - Aman Chandra Kaushik
- The State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Dong Qing Wei
- The State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Center of Interdisciplinary Sciences-Computational Life Sciences, Henan University of Technology, Zhengzhou, China
- Peng Cheng Laboratory, Shenzhen, China
| |
Collapse
|
28
|
Guo Z, Huang J, Wang Y, Liu XP, Li W, Yao J, Li S, Hu W. Analysis of Expression and Its Clinical Significance of the Secreted Phosphoprotein 1 in Lung Adenocarcinoma. Front Genet 2020; 11:547. [PMID: 32595698 PMCID: PMC7303289 DOI: 10.3389/fgene.2020.00547] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Objective To explore the expression of secreted phosphoprotein 1 (SPP1) in lung adenocarcinoma (LUAD), and evaluate its relationship with clinicopathological characteristics and prognosis of LUAD, and analyze the advantages of SPP1 as a potential prognostic marker in LUAD. Methods The expression of SPP1 in normal lung tissue and LUAD was analyzed from the Cancer Cell Line Encyclopedia (CCLE), Gene Expression Omnibus (GEO), and Human Protein Atlas (HPA) databases. GSE68465 was used to explore the relationship between the SPP1 expression and clinicopathological characteristics and the prognosis of LUAD patients. The relationship between SPP1 and immune infiltration in LUAD was analyzed by the Tumor Immune Estimation Resource (TIMER) database. Gene enrichment analysis was performed in GSEA. The Cancer Genome Atlas (TCGA)-LUAD data was used to verify the results. Results In the cell line level, non-small cell lung cancer ranked ninth among cancer cell lines based on SPP1 expression. In the messenger RNA (mRNA) and protein levels, SPP1 expression was higher in LUAD tissues than that in normal control. SPP1 expression was related to gender, N stage, histological grade, and progression or relapse. In men, SPP1 expression were higher compared to that in women. The higher the N stage, the higher the SPP1 expression level. As LUAD progresses or relapses, SPP1 expression could increase. In the pathological grade, the SPP1 expression was higher in LUAD samples with moderate differentiation. In addition, the overall 5-year survival rates of the SPP1 high and low expression groups were 50.574 and 59.181% [P = 0.008; hazard ratio (HR) = 0.7057; 95% CI, 0.5467-0.9109], indicating that SPP1 had an impact on overall survival for LUAD patients. The relationship between SPP1 expression and CD4+ T cell, macrophage, neutrophil, and dendritic cell infiltration was weak in LUAD. SPP1 could be considered as an independent prognostic marker in LUAD (P = 0.003; HR = 1.150; 95% CI, 1.048-1.261) by multivariate Cox regression analysis. The results of GSEA indicated that samples with high SPP1 expression were enriched in protein secretion, mTORC1 signaling, angiogenesis, and glycolysis pathway. The analysis results obtained by TCGA-LUAD data were basically consistent with the results obtained by GSE68465. Conclusions SPP1 can not only affect the occurrence and development of LUAD but also may be an independent prognostic marker of LUAD. SPP1 is expected to be a new target for molecular targeted therapy.
Collapse
Affiliation(s)
- Zixin Guo
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingyu Huang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yujin Wang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiao-Ping Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Li
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, China
| | - Jie Yao
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Sheng Li
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China
| | - Weidong Hu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Li M, Wang K, Pang Y, Zhang H, Peng H, Shi Q, Zhang Z, Cui X, Li F. Secreted Phosphoprotein 1 (SPP1) and Fibronectin 1 (FN1) Are Associated with Progression and Prognosis of Esophageal Cancer as Identified by Integrated Expression Profiles Analysis. Med Sci Monit 2020; 26:e920355. [PMID: 32208405 PMCID: PMC7111131 DOI: 10.12659/msm.920355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Esophageal cancer is a malignant tumor with a complex pathogenesis and a poor 5-year survival rate, which encourages researchers to explore its molecular mechanisms deeper to improve the prognosis. Material/Methods DEGs were from 4 Gene Expression Omnibus (GEO) databases (GSE92396, GSE20347, GSE23400, and GSE45168) including 87 esophageal tumor samples and 84 normal samples. We performed Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Protein-Protein interaction (PPI) analysis, and GeneMANIA to identify the DEGs. Gene set enrichment analysis (GSEA) and Kaplan-Meier survival analyses were performed. Results There was an overlapping subset consisting of 120 DEGs that was present in all esophageal tumor samples. The DEGs were enriched in extracellular matrix (ECM)-receptor interaction, as well as focal adhesion and transcriptional mis-regulation in cancer. The 2 most crucial regulatory pathways in esophageal cancer were the amebiasis pathway and the PI3K-Akt signaling pathway. Secreted phosphoprotein 1 (SPP1) and fibronectin 1 (FN1) were selected and verified in an independent cohort and samples using the TCGA and GTEx projects. Gene set enrichment analysis (GSEA) showed that proteasome and nucleotide excision repair were 2 most differentially enriched pathways in the SPP1 high-expression phenotype, and ECM-receptor interaction and focal adhesion in FN1 high-expression phenotype. Kaplan-Meier survival analysis showed that SPP1 and FN1 were significantly positively related to overall survival and had the potential to predict patient relapse. Conclusions Our analysis is the first to show that SPP1 and FN1 might work as biological markers of progression and prognosis in esophageal carcinoma (ESCA).
Collapse
Affiliation(s)
- Menglu Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China (mainland)
| | - Kaige Wang
- Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Yanhua Pang
- Department of Gastroenterology, Beijing Chaoyang Hospital, Beijing, China (mainland)
| | - Hongpan Zhang
- Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Hao Peng
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China (mainland)
| | - Qi Shi
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China (mainland)
| | - Zhiyu Zhang
- Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Xiaobin Cui
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China (mainland)
| | - Feng Li
- Department of Pathology and Key Laboratory for Xinjiang Endemic and Ethnic Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China (mainland).,Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| |
Collapse
|
30
|
Esophageal Cancer Development: Crucial Clues Arising from the Extracellular Matrix. Cells 2020; 9:cells9020455. [PMID: 32079295 PMCID: PMC7072790 DOI: 10.3390/cells9020455] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
In the last years, the extracellular matrix (ECM) has been reported as playing a relevant role in esophageal cancer (EC) development, with this compartment being related to several aspects of EC genesis and progression. This sounds very interesting due to the complexity of this highly incident and lethal tumor, which takes the sixth position in mortality among all tumor types worldwide. The well-established increase in ECM stiffness, which is able to trigger mechanotransduction signaling, is capable of regulating several malignant behaviors by converting alteration in ECM mechanics into cytoplasmatic biochemical signals. In this sense, it has been shown that some molecules play a key role in these events, particularly the different collagen isoforms, as well as enzymes related to its turnover, such as lysyl oxidase (LOX) and matrix metalloproteinases (MMPs). In fact, MMPs are not only involved in ECM stiffness, but also in other events related to ECM homeostasis, which includes ECM remodeling. Therefore, the crucial role of distinct MMPs isoform has already been reported, especially MMP-2, -3, -7, and -9, along EC development, thus strongly associating these proteins with the control of important cellular events during tumor progression, particularly in the process of invasion during metastasis establishment. In addition, by distinct mechanisms, a vast diversity of glycoproteins and proteoglycans, such as laminin, fibronectin, tenascin C, galectin, dermatan sulfate, and hyaluronic acid exert remarkable effects in esophageal malignant cells due to the activation of oncogenic signaling pathways mainly involved in cytoskeleton alterations during adhesion and migration processes. Finally, the wide spectrum of interactions potentially mediated by ECM may represent a singular intervention scenario in esophageal carcinogenesis natural history and, due to the scarce knowledge on the cellular and molecular mechanisms involved in EC development, the growing body of evidence on ECM’s role along esophageal carcinogenesis might provide a solid base to improve its management in the future.
Collapse
|
31
|
Kovács D, Igaz N, Marton A, Rónavári A, Bélteky P, Bodai L, Spengler G, Tiszlavicz L, Rázga Z, Hegyi P, Vizler C, Boros IM, Kónya Z, Kiricsi M. Core-shell nanoparticles suppress metastasis and modify the tumour-supportive activity of cancer-associated fibroblasts. J Nanobiotechnology 2020; 18:18. [PMID: 31964403 PMCID: PMC6974972 DOI: 10.1186/s12951-020-0576-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Although accumulating evidence suggests that the crosstalk between malignant cells and cancer-associated fibroblasts (CAFs) actively contributes to tumour growth and metastatic dissemination, therapeutic strategies targeting tumour stroma are still not common in the clinical practice. Metal-based nanomaterials have been shown to exert excellent cytotoxic and anti-cancerous activities, however, their effects on the reactive stroma have never been investigated in details. Thus, using feasible in vitro and in vivo systems to model tumour microenvironment, we tested whether the presence of gold, silver or gold-core silver-shell nanoparticles exerts anti-tumour and metastasis suppressing activities by influencing the tumour-supporting activity of stromal fibroblasts. RESULTS We found that the presence of gold-core silver-shell hybrid nanomaterials in the tumour microenvironment attenuated the tumour cell-promoting behaviour of CAFs, and this phenomenon led to a prominent attenuation of metastatic dissemination in vivo as well. Mechanistically, transcriptome analysis on tumour-promoting CAFs revealed that silver-based nanomaterials trigger expressional changes in genes related to cancer invasion and tumour metastasis. CONCLUSIONS Here we report that metal nanoparticles can influence the cancer-promoting activity of tumour stroma by affecting the gene expressional and secretory profiles of stromal fibroblasts and thereby altering their intrinsic crosstalk with malignant cells. This potential of metal nanomaterials should be exploited in multimodal treatment approaches and translated into improved therapeutic outcomes.
Collapse
Affiliation(s)
- Dávid Kovács
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary
| | - Annamária Marton
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Andrea Rónavári
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary
- Interdisciplinary Excellence Centre, Department of Applied and Environmental Chemistry, University of Szeged, Rerrich B. tér 1, 6720, Szeged, Hungary
| | - Péter Bélteky
- Interdisciplinary Excellence Centre, Department of Applied and Environmental Chemistry, University of Szeged, Rerrich B. tér 1, 6720, Szeged, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary
| | - Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Dóm tér 9, 6720, Szeged, Hungary
| | - László Tiszlavicz
- Department of Pathology, University of Szeged, Állomás u. 2, 6725, Szeged, Hungary
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, Állomás u. 2, 6725, Szeged, Hungary
| | - Péter Hegyi
- First Department of Medicine, University of Szeged, Korányi fasor 8-10, 6720, Szeged, Hungary
- MTA-SZTE Lendület Translational Gastroenterology Research Group, Korányi fasor 8-10, 6720, Szeged, Hungary
| | - Csaba Vizler
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Imre M Boros
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary
- Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Temesvári Krt. 62, 6726, Szeged, Hungary
| | - Zoltán Kónya
- Interdisciplinary Excellence Centre, Department of Applied and Environmental Chemistry, University of Szeged, Rerrich B. tér 1, 6720, Szeged, Hungary
- MTA-SZTE Reaction Kinetics and Surface Chemistry Research Group, Rerrich B. tér 1, 6720, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52, 6726, Szeged, Hungary.
| |
Collapse
|
32
|
Integrated Bioinformatics Analysis Identifies Hub Genes Associated with the Pathogenesis and Prognosis of Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2615921. [PMID: 31950035 PMCID: PMC6948276 DOI: 10.1155/2019/2615921] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) accounts for over 90% of all esophageal tumors. However, the molecular mechanism underlying ESCC development and prognosis remains unclear, and there are still no effective molecular biomarkers for diagnosing or predicting the clinical outcome of patients with ESCC. Here, using bioinformatics analyses, we attempted to identify potential biomarkers and therapeutic targets for ESCC. Differentially expressed genes (DEGs) between ESCC and normal esophageal tissue samples were obtained through comprehensive analysis of three publicly available gene expression profile datasets from the Gene Expression Omnibus database. The biological roles of the DEGs were identified by Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. Moreover, the Cytoscape 3.7.1 platform and subsidiary tools such as Molecular Complex Detection (MCODE) and CytoHubba were used to visualize the protein-protein interaction (PPI) network of the DEGs and identify hub genes. A total of 345 DEGs were identified between normal esophageal and ESCC samples, which were enriched in the KEGG pathways of the cell cycle, endocytosis, pancreatic secretion, and fatty acid metabolism. Two of the highest scoring models were selected from the PPI network using Molecular Complex Detection. Moreover, CytoHubba revealed 21 hub genes with a valuable influence on the progression of ESCC in these patients. Among these, the high expression levels of five genes-SPP1, SPARC, BGN, POSTN, and COL1A2-were associated with poor disease-free survival of ESCC patients, as indicated by survival analysis. Taken together, we identified that elevated expression of five hub genes, including SPP1, is associated with poor prognosis in ESCC patients, which may serve as potential prognostic biomarkers or therapeutic target for ESCC.
Collapse
|
33
|
Barranco G, Fernández E, Rivas S, Quezada R, Nava D, Aguilar J, García A, Astudillo H, Lome C, Ruiz E. Osteopontin expression and its relationship with prognostic factors in diffuse large B-cell lymphoma. Hematol Rep 2019; 11:7964. [PMID: 31579151 PMCID: PMC6761465 DOI: 10.4081/hr.2019.7964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 07/05/2019] [Indexed: 01/20/2023] Open
Abstract
The aim of this study is to explore the expression of osteopontin (OPN) and its relationship with prognostic factors and survival in diffuse large B cell lymphoma (DLBCL). A tissue microarray was performed for immunohistochemical evaluation. Contingency tables were analyzed for trends; chi-square test was used to determine differences between groups. Univariate and multivariate Cox proportional hazards-regression analyses were performed to evaluate the impact of prognostic factors on survival. Expression of OPN was observed in 28%. It was different in non-germinal center DLBCL (P=0.04). The mean overall survival (OS) was lower in patients with positive OPN expression (19.762; CI 95% 14.269-25.255) it was not significant (P=0.123). It is not possible to establish a clear relationship between the expression by immunohistochemistry of osteopontin and a poor prognosis but it would be important to complement the analysis with other techniques as PCR or NGS that allow us to assess the influence of the isoforms and post-translational modifications of OPN on the biological behavior of DLBCL. Our findings indicate that OPN expression could be associated with a more aggressive variant of lymphoma: non-germinal center DLBCL.
Collapse
Affiliation(s)
| | - Edith Fernández
- Translational Medicine, National Institute of Cancerology, Mexico City.,Computational Genomics, National Institute of Genomic Medicine, Mexico City
| | - Silvia Rivas
- Department of Hematology, National Institute of Cancerology, Mexico City
| | - Roxana Quezada
- Department of Pathology, National Institute of Cancerology, Mexico City
| | - Dolores Nava
- Department of Pathology, National Institute of Cancerology, Mexico City
| | - José Aguilar
- Department of Internal Medicine, National Institute of Cancerology, Mexico City
| | - Abelardo García
- Translational Medicine, National Institute of Cancerology, Mexico City
| | - Horacio Astudillo
- Laboratory of Translational Cancer Research and Cellular Therapy, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Carmen Lome
- Department of Pathology, National Institute of Cancerology, Mexico City
| | - Erika Ruiz
- Translational Medicine, National Institute of Cancerology, Mexico City
| |
Collapse
|
34
|
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
|
35
|
Cai C, Cooper GF, Lu KN, Ma X, Xu S, Zhao Z, Chen X, Xue Y, Lee AV, Clark N, Chen V, Lu S, Chen L, Yu L, Hochheiser HS, Jiang X, Wang QJ, Lu X. Systematic discovery of the functional impact of somatic genome alterations in individual tumors through tumor-specific causal inference. PLoS Comput Biol 2019; 15:e1007088. [PMID: 31276486 PMCID: PMC6650088 DOI: 10.1371/journal.pcbi.1007088] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/23/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer is mainly caused by somatic genome alterations (SGAs). Precision oncology involves identifying and targeting tumor-specific aberrations resulting from causative SGAs. We developed a novel tumor-specific computational framework that finds the likely causative SGAs in an individual tumor and estimates their impact on oncogenic processes, which suggests the disease mechanisms that are acting in that tumor. This information can be used to guide precision oncology. We report a tumor-specific causal inference (TCI) framework, which estimates causative SGAs by modeling causal relationships between SGAs and molecular phenotypes (e.g., transcriptomic, proteomic, or metabolomic changes) within an individual tumor. We applied the TCI algorithm to tumors from The Cancer Genome Atlas (TCGA) and estimated for each tumor the SGAs that causally regulate the differentially expressed genes (DEGs) in that tumor. Overall, TCI identified 634 SGAs that are predicted to cause cancer-related DEGs in a significant number of tumors, including most of the previously known drivers and many novel candidate cancer drivers. The inferred causal relationships are statistically robust and biologically sensible, and multiple lines of experimental evidence support the predicted functional impact of both the well-known and the novel candidate drivers that are predicted by TCI. TCI provides a unified framework that integrates multiple types of SGAs and molecular phenotypes to estimate which genome perturbations are causally influencing one or more molecular/cellular phenotypes in an individual tumor. By identifying major candidate drivers and revealing their functional impact in an individual tumor, TCI sheds light on the disease mechanisms of that tumor, which can serve to advance our basic knowledge of cancer biology and to support precision oncology that provides tailored treatment of individual tumors.
Collapse
Affiliation(s)
- Chunhui Cai
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Gregory F. Cooper
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Kevin N. Lu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Xiaojun Ma
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Shuping Xu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Zhenlong Zhao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xueer Chen
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Yifan Xue
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Adrian V. Lee
- Center for Causal Discovery, Pittsburgh, PA, United States of America
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
- Magee Women’s Cancer Research Center, Pittsburgh, PA, United States of America
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
| | - Nathan Clark
- Center for Causal Discovery, Pittsburgh, PA, United States of America
- Department of Computational Biology and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Vicky Chen
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Songjian Lu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Lujia Chen
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Liyue Yu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Harry S. Hochheiser
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Xia Jiang
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
| | - Q. Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail: (QJW); (XL)
| | - Xinghua Lu
- Department of Biomedical Informatics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Causal Discovery, Pittsburgh, PA, United States of America
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States of America
- * E-mail: (QJW); (XL)
| |
Collapse
|
36
|
Hai L, Szwarc MM, Lonard DM, Rajapakshe K, Perera D, Coarfa C, Ittmann M, Fernandez-Valdivia R, Lydon JP. Short-term RANKL exposure initiates a neoplastic transcriptional program in the basal epithelium of the murine salivary gland. Cytokine 2019; 123:154745. [PMID: 31226438 DOI: 10.1016/j.cyto.2019.154745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022]
Abstract
Although salivary gland cancers comprise only ∼3-6% of head and neck cancers, treatment options for patients with advanced-stage disease are limited. Because of their rarity, salivary gland malignancies are understudied compared to other exocrine tissue cancers. The comparative lack of progress in this cancer field is particularly evident when it comes to our incomplete understanding of the key molecular signals that are causal for the development and/or progression of salivary gland cancers. Using a novel conditional transgenic mouse (K5:RANKL), we demonstrate that Receptor Activator of NFkB Ligand (RANKL) targeted to cytokeratin 5-positive basal epithelial cells of the salivary gland causes aggressive tumorigenesis within a short period of RANKL exposure. Genome-wide transcriptomic analysis reveals that RANKL markedly increases the expression levels of numerous gene families involved in cellular proliferation, migration, and intra- and extra-tumoral communication. Importantly, cross-species comparison of the K5:RANKL transcriptomic dataset with The Cancer Genome Atlas cancer signatures reveals the strongest molecular similarity with cancer subtypes of the human head and neck squamous cell carcinoma. These studies not only provide a much needed transcriptomic resource to mine for novel molecular targets for therapy and/or diagnosis but validates the K5:RANKL transgenic as a preclinical model to further investigate the in vivo oncogenic role of RANKL signaling in salivary gland tumorigenesis.
Collapse
Affiliation(s)
- Lan Hai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou, Henan Province, PR China
| | - Maria M Szwarc
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Dimuthu Perera
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael Ittmann
- Department of Pathology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | | | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
37
|
Osteopontin isoforms differentially promote arteriogenesis in response to ischemia via macrophage accumulation and survival. J Transl Med 2019; 99:331-345. [PMID: 29959420 PMCID: PMC6311150 DOI: 10.1038/s41374-018-0094-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/16/2018] [Accepted: 05/29/2018] [Indexed: 01/13/2023] Open
Abstract
Osteopontin (OPN) is critical for ischemia-induced neovascularization. Unlike rodents, humans express three OPN isoforms (a, b, and c); however, the roles of these isoforms in post-ischemic neovascularization and cell migration remain undefined. Our objective was to determine if OPN isoforms differentially affect post-ischemic neovascularization and to elucidate the mechanisms underlying these differences. To investigate if human OPN isoforms exert divergent effects on post-ischemic neovascularization, we utilized OPN-/- mice and a loss-of-function/gain-of-function approach in vivo and in vitro. In this study OPN-/- mice underwent hindlimb ischemia surgery and 1.5 × 106 lentivirus particles were administered intramuscularly to overexpress OPNa, OPNb, or OPNc. OPNa and OPNc significantly improved limb perfusion 30.4% ± 0.8 and 70.9% ± 6.3, respectively, and this translated to improved functional limb use, as measured by voluntary running wheel utilization. OPNa- and OPNc-treated animals exhibited significant increases in arteriogenesis, defined here as the remodeling of existing arterioles into larger conductance arteries. Macrophages play a prominent role in the arteriogenesis process and OPNa- and OPNc-treated animals showed significant increases in macrophage accumulation in vivo. In vitro, OPN isoforms did not affect macrophage polarization, whereas all three isoforms increased macrophage survival and decreased macrophage apoptosis. However, OPN isoforms exert differential effects on macrophage migration, where OPNa and OPNc significantly increased macrophage migration, with OPNc serving as the most potent isoform. In conclusion, human OPN isoforms exert divergent effects on neovascularization through differential effects on arteriogenesis and macrophage accumulation in vivo and on macrophage migration and survival, but not polarization, in vitro. Altogether, these data support that human OPN isoforms may represent novel therapeutic targets to improve neovascualrization and preserve tissue function in patients with obstructive artery diseases.
Collapse
|
38
|
Han X, Wang W, He J, Jiang L, Li X. Osteopontin as a biomarker for osteosarcoma therapy and prognosis. Oncol Lett 2019; 17:2592-2598. [PMID: 30854034 PMCID: PMC6365895 DOI: 10.3892/ol.2019.9905] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone malignancy, and is particularly prevalent in children and adolescents. OS is an aggressive tumor with a tendency to metastasize and invade to para-carcinoma tissues. The primary treatment for this tumor is a combination of surgery and chemotherapy. However, the prognosis remains poor due to chemoresistance and early metastasis. Osteopontin (OPN), a multifunctional secreted protein, has emerged as an important potential biomarker for diagnosing and treating cancer. The overexpression of OPN has been found in numerous malignant tumors, including breast, lung, gastric and ovarian cancer, as well as melanoma. Recent studies have suggested that OPN may provide an important function in the diagnosis and treatment of OS. The present review summarizes current knowledge and progress in understanding the potential role of OPN as a biomarker in OS.
Collapse
Affiliation(s)
- Xingwen Han
- Department of Orthopedics, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wenji Wang
- Department of Orthopedics, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jingjing He
- Department of Liver Diseases, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Lei Jiang
- Department of Oncology Surgery, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xun Li
- Department of General Surgery, First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
39
|
Yang H, Wu J, Zhang J, Yang Z, Jin W, Li Y, Jin L, Yin L, Liu H, Wang Z. Integrated bioinformatics analysis of key genes involved in progress of colon cancer. Mol Genet Genomic Med 2019; 7:e00588. [PMID: 30746900 PMCID: PMC6465657 DOI: 10.1002/mgg3.588] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/21/2018] [Accepted: 01/06/2019] [Indexed: 12/26/2022] Open
Abstract
Background Colon cancer is one of most malignant cancers around worldwide. Nearly 20% patients were diagnosed at colon cancer with metastasis. However, the lack of understanding regarding its pathogenesis brings difficulties to study it. Methods In this study, we acquired high‐sequence data from GEO dataset, and performed integrated bioinformatic analysis including differently expressed genes, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathways analysis, protein–protein analysis, survival analysis to analyze the development of colon cancer. Results By comparing the colon cancer tissues with normal colon tissues, 109 genes were dysregulated; among them, 83 genes were downregulated and 26 genes were upregulated. Two clusters were founded based on the STRING database and MCODE plugin of cytoscape software. Then, six genes with prognostic value were filtered out in UALCAN website. Conclusion We found that SPP1, VIP, COL11A1, CA2, ADAM12, INHBA could provide great significant prognostic value for colon cancer.
Collapse
Affiliation(s)
- Haojie Yang
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiong Wu
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Zhang
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhigang Yang
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Jin
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Li
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Jin
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Yin
- Shanghai Changning Maternity & Infant Health Hospital, Shanghai, China
| | - Hua Liu
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhenyi Wang
- Department of colo-proctology, Yueyang Hospital of Integratd Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Gimba E, Brum M, Nestal De Moraes G. Full-length osteopontin and its splice variants as modulators of chemoresistance and radioresistance (Review). Int J Oncol 2018; 54:420-430. [DOI: 10.3892/ijo.2018.4656] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/25/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Etel Gimba
- Program of Cellular and Molecular Oncobiology, National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | - Mariana Brum
- Program of Cellular and Molecular Oncobiology, National Cancer Institute, Rio de Janeiro 20231-050, Brazil
| | - Gabriela Nestal De Moraes
- Cellular and Molecular Hemato-Oncology Laboratory, Molecular Hemato-Oncology Program, National Cancer Institute, Rio de Janeiro 20230-130, Brazil
| |
Collapse
|
41
|
Xu Y, Shen K. Identification of potential key genes associated with ovarian clear cell carcinoma. Cancer Manag Res 2018; 10:5461-5470. [PMID: 30519094 PMCID: PMC6234989 DOI: 10.2147/cmar.s187156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Ovarian cancer is the major cause of death from cancer among females worldwide. Ovarian clear cell carcinoma (OCCC) is considered a distinct histopathologic subtype with worse prognosis and resistance to conventional chemotherapy. Materials and methods We analyzed five microarray datasets derived from the Gene Expression Omnibus database. GEO2R tool was used to screen out differentially expressed genes (DEGs) between OCCC tumor and normal ovary tissue. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were performed using the g:Profiler database and Cytoscape. Based on Search Tool for the Retrieval of Interacting Genes, we performed protein-protein interaction (PPI) network analysis on the DEGs. Real-time PCR (RT-PCR) and Western blotting in frozen samples of normal ovary and OCCC were performed to verify the expression difference of hub genes in OCCC patients. Results Thirty upregulated DEGs and 13 downregulated DEGs were identified by cross referencing. Six were chosen as hub genes with high connectivity degree via PPI network analysis, including two upregulated and four downregulated. RT-PCR and Western blotting results showed significant expression difference of the two upregulated genes, SPP1 and EPCAM, between tumor and normal tissues. Conclusion Our research suggests that SPP1 and EPCAM are overexpressed in OCCC compared with normal ovary tissue. Clinical study of large sample is required to evaluate the value of SPP1 and EPCAM in the precision treatment and prognostic influence on OCCC in the future.
Collapse
Affiliation(s)
- Youzheng Xu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Eastern District, Beijing 100730, China,
| | - Keng Shen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Eastern District, Beijing 100730, China,
| |
Collapse
|
42
|
He F, Ai B, Tian L. Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis. Biomed Rep 2018; 9:305-312. [PMID: 30233782 PMCID: PMC6142036 DOI: 10.3892/br.2018.1134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/19/2018] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is one of the most common subtypes of esophageal cancer, and is associated with a low 5-year survival rate. The present study aimed to identify key genes and pathways associated with EAC using bioinformatics analysis. The gene expression profiles of GSE92396, which includes 12 EAC samples and 9 normal esophageal samples, were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between the EAC and normal samples were identified using the limma package in R language. Gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the identified DEGs were conducted using the online analysis tool, the Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction (PPI) network of the DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes (STRING) database and Cytoscape software. Finally, module analysis was conducted for the PPI network using the MCODE plug-in in Cytoscape. Of the 386 DEGs identified, the 150 upregulated genes were mainly enriched in the KEGG pathways of complement and coagulation cascades, maturity onset diabetes of the young and protein digestion and absorption; and the 236 downregulated genes were mainly enriched in amoebiasis, retinol metabolism and drug metabolism-cytochrome P450. Based on information from the STRING database, a PPI network comprising of 369 nodes and 534 edges was constructed in Cytoscape. The top 10 hub nodes with the highest degrees were determined as interleukin-8, involucrin, tissue inhibitor of metalloproteinase 1, fibronectin 1, serpin family E member 1, serpin family A member 1, cystic fibrosis transmembrane conductance regulator, secreted phosphoprotein 1, collagen type I alpha 1 chain and angiotensinogen. A total of 6 modules were detected from the PPI network that satisfied the criteria of MCODE score >4 and number of nodes >4. KEGG pathways enriched for the module DEGs were mainly within arachidonic acid metabolism, complement and coagulation cascades and rheumatoid arthritis. In conclusion, identification of these key genes and pathways may improve understanding of the mechanisms underlying the development of EAC, and may be used as diagnostic and therapeutic targets in EAC.
Collapse
Affiliation(s)
- Feng He
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Bo Ai
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Lei Tian
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
43
|
Abstract
Cancer metastasis is defined as the dissemination of malignant cells from the primary tumor site, leading to colonization of distant organs and the establishment of a secondary tumor. Metastasis is frequently associated with chemoresistance and is the major cause of cancer-related mortality. Metastatic cells need to acquire the ability to resist to stresses provided by different environments, such as reactive oxygen species, shear stress, hemodynamic forces, stromal composition, and immune responses, to colonize other tissues. Hence, only a small population of cells has a metastasis-initiating potential. Several studies have revealed the misregulation of transcriptional variants during cancer progression, and many splice events can be used to distinguish between normal and tumoral tissue. These variants, which are abnormally expressed in malignant cells, contribute to an adaptive response of tumor cells and the success of the metastatic cascade, promoting an anomalous cell cycle, cellular adhesion, resistance to death, cell survival, migration and invasion. Understanding the different aspects of splicing regulation and the influence of transcriptional variants that control metastatic cells is critical for the development of therapeutic strategies. In this review, we describe how transcriptional variants contribute to metastatic competence and discuss how targeting specific isoforms may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Joice De Faria Poloni
- a Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| | - Diego Bonatto
- a Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia , Universidade Federal do Rio Grande do Sul , Porto Alegre , RS , Brazil
| |
Collapse
|
44
|
Zeng B, Zhou M, Wu H, Xiong Z. SPP1 promotes ovarian cancer progression via Integrin β1/FAK/AKT signaling pathway. Onco Targets Ther 2018; 11:1333-1343. [PMID: 29559792 PMCID: PMC5856063 DOI: 10.2147/ott.s154215] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES Ovarian cancer is one of the most lethal malignant tumors in women. Secreted phosphoprotein 1 (SPP1) plays an important role in some cancer types. Therefore, the role of SPP1 in ovarian cancer was determined and the potential mechanism was elucidated. MATERIALS AND METHODS The expression of SPP1 in ovarian cancer was determined by immunohistochemistry in ovarian cancer tissues and normal ovarian tissues. Cellular proliferation, migration, and invasion were determined by cell counting kit-8 assay, wound healing assay, and Matrigel invasion assay in SKOV3 and A2780 cells. The protein expression of SPP1, integrin subunit β1 (Integrin β1), focal adhesion kinase (FAK), and phosphorylation protein kinase B (p-AKT) was detected by Western blotting in SKOV3 cells after silencing SPP1. The expression of SPP1 was determined in SKOV3 cells after transfecting with miR-181a mimics or inhibitors. The growth of SKOV3 cells in vivo was determined in a nude mouse model of ovarian cancer after silencing SPP1. RESULTS The expression of SPP1 was higher in epithelial ovarian cancer tissues than in normal ovarian tissues. Silencing SPP1 decreased the cell proliferation, migration, and invasion. Ectopic expression of SPP1 increased the cell proliferation, migration, and invasion. Silencing SPP1 prevented ovarian cancer growth in mice. Silencing SPP1 inhibited Integrin β1/FAK/AKT pathway. In agreement, ectopically expressed SPP1 activated Integrin β1/FAK/AKT pathway. Also, SPP1 was regulated by miR-181a. CONCLUSION SPP1 is a biomarker for the prognosis of ovarian cancer. It is also oncogenic and a potential target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Biao Zeng
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Min Zhou
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Huan Wu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhengai Xiong
- Department of Obstetrics and Gynecology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
45
|
Osteopontin-a splice variant is overexpressed in papillary thyroid carcinoma and modulates invasive behavior. Oncotarget 2018; 7:52003-52016. [PMID: 27409830 PMCID: PMC5239531 DOI: 10.18632/oncotarget.10468] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/18/2016] [Indexed: 01/04/2023] Open
Abstract
Osteopontin (OPN) is a matricellular protein overexpressed in cancer cells and modulates tumorigenesis and metastasis, including in thyroid cancer (TC). The contribution of each OPN splice variant (OPN-SV), named OPNa, OPNb and OPNc, in TC is currently unknown. This study evaluates the expression of total OPN (tOPN) and OPN-SV in TC tissues and cell lines, their correlation with clinicopathological, molecular features and their functional roles. We showed that tOPN and OPNa are overexpressed in classic papillary thyroid carcinoma (cPTC) in relation to adjacent thyroid, adenoma and follicular variant of papillary thyroid carcinoma (fvPTC) tissues. In cPTC, OPNa overexpression is associated with larger tumor size, vascular invasion, extrathyroid extension and BRAFV600E mutation. We found that TC cell lines overexpressing OPNa exhibited increased proliferation, migration, motility and in vivo invasion. Conditioned medium secreted from cells overexpressing OPNa induce MMP2 and MMP9 metalloproteinases activity. In summary, we described the expression pattern of OPN-SV in cPTC samples and the key role of OPNa expression on activating TC tumor progression features. Our findings highlight OPNa variant as TC biomarker, besides being a putative target for cPTC therapeutic approaches.
Collapse
|
46
|
Is Osteopontin a Friend or Foe of Cell Apoptosis in Inflammatory Gastrointestinal and Liver Diseases? Int J Mol Sci 2017; 19:ijms19010007. [PMID: 29267211 PMCID: PMC5795959 DOI: 10.3390/ijms19010007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
Osteopontin (OPN) is involved in a variety of biological processes, including bone remodeling, innate immunity, acute and chronic inflammation, and cancer. The expression of OPN occurs in various tissues and cells, including intestinal epithelial cells and immune cells such as macrophages, dendritic cells, and T lymphocytes. OPN plays an important role in the efficient development of T helper 1 immune responses and cell survival by inhibiting apoptosis. The association of OPN with apoptosis has been investigated. In this review, we described the role of OPN in inflammatory gastrointestinal and liver diseases, focusing on the association of OPN with apoptosis. OPN changes its association with apoptosis depending on the type of disease and the phase of disease activity, acting as a promoter or a suppressor of inflammation and inflammatory carcinogenesis. It is essential that the roles of OPN in those diseases are elucidated, and treatments based on its mechanism are developed.
Collapse
|
47
|
Yue Y, Song M, Qiao Y, Li P, Yuan Y, Lian J, Wang S, Zhang Y. Gene function analysis and underlying mechanism of esophagus cancer based on microarray gene expression profiling. Oncotarget 2017; 8:105222-105237. [PMID: 29285246 PMCID: PMC5739633 DOI: 10.18632/oncotarget.22160] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/28/2017] [Indexed: 12/20/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common digestive malignant tumors worldwide. Over the past decades, there have been minimal improvements in outcomes for patients with EC. New targets and novel therapies are needed to improve outcomes for these patients. This study aimed to explore the molecular mechanisms of EC by integrated bioinformatic analyses of the feature genes associated with EC and correlative gene functions which can distinguish cancerous tissues from non-cancerous tissues. Gene expression profile GSE20347 was downloaded from Gene Expression Omnibus (GEO) database, including 17 EC samples and their paired adjacent non-cancerous samples. The differentially expressed genes (DEGs) between EC and normal specimens were identified and then applied to analyze the GO enrichment on gene functions and KEGG pathways. Corresponding Pathway Relation Network (Pathway-net) and Gene Signal Network (signal-net) of DEGs were established based on the data collected from GCBI datasets. The results showed that DEGs mainly participated in the process of cell adhesion, cell proliferation, survival, invasion, metastasis and angiogenesis. Aberrant expression of PTK2, MAPK signaling pathway, PI3K-Akt signaling pathway, p53 signaling pathway and MET were closely associated with EC carcinogenesis. Importantly, Interleukin 8 (IL8) and C-X-C chemokine receptor type 7 (CXCR-7) were predicted to be significantly related to EC. These findings were further validated by analyzing both TCGA database and our clinical samples of EC. Our discovery provides a registry of genes and pathways that are disrupted in EC, which has the potential to be used in clinic for diagnosis and target therapy of EC in future.
Collapse
Affiliation(s)
- Ying Yue
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- The No.7. People's Hospital of Zhengzhou, Zhengzhou, Henan 450016, China
| | - Mengjia Song
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yamin Qiao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Pupu Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yiqiang Yuan
- The No.7. People's Hospital of Zhengzhou, Zhengzhou, Henan 450016, China
| | - Jingyao Lian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Suying Wang
- Clinical Laboratory, Hebi People's Hospital, Hebi 458030, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- Key Laboratory for Tumor Immunology and Biotherapy of Henan Province, Zhengzhou, Henan 450052, China
| |
Collapse
|
48
|
Wong JP, Wei R, Lyu P, Tong OL, Zhang SD, Wen Q, Yuen HF, El-Tanani M, Kwok HF. Clinical and in vitro analysis of Osteopontin as a prognostic indicator and unveil its potential downstream targets in bladder cancer. Int J Biol Sci 2017; 13:1373-1386. [PMID: 29209142 PMCID: PMC5715521 DOI: 10.7150/ijbs.21457] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Osteopontin (OPN) plays an important role in cancer progression, however its prognostic significance and its downstream factors are largely elusive. In this study, we have shown that expression of OPN was significantly higher in bladder cancer specimens with higher T-stage or tumor grades. In addition, a high level of OPN was significantly associated with poorer survival in two independent bladder cancer patient cohorts totaling 389 bladder cancer patients with available survival data. We further identified Matrix metallopeptidase 9 (MMP9) and S100 calcium-binding protein A8 (S100A8) were both downstream factors for OPN in bladder cancer specimens and bladder cancer cell lines. Expression of OPN was significantly positively associated with that of MMP9 and S100A8, while overexpression of OPN resulted in upregulation of MMP9 and S100A8, and knockdown of OPN showed consistent downregulation of MMP9 and S100A8 expression levels. Importantly, expression levels of both MMP9 and S100A8 were significantly associated with higher T-stage, higher tumor grade and a shorter survival time in the bladder cancer patients. Interestingly, OPN expression only predicted survival in MMP9-high, but not MMP9-low subgroups, and in S100A8-low but not S100A8-high subgroups. Our results suggest that OPN, MMP9 and S100A8 all play a significant role in bladder cancer progression and are potential prognostic markers and therapeutic targets in bladder cancer. The mechanistic link between these three genes and bladder cancer progression warrants further investigation.
Collapse
Affiliation(s)
- Janet P.C. Wong
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Ran Wei
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Peng Lyu
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Olivia L.H. Tong
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Shu Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Londonderry, United Kingdom
| | - Qing Wen
- Center for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, United Kingdom
| | - Hiu Fung Yuen
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Mohamed El-Tanani
- Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| |
Collapse
|
49
|
Wei R, Wong JPC, Kwok HF. Osteopontin -- a promising biomarker for cancer therapy. J Cancer 2017; 8:2173-2183. [PMID: 28819419 PMCID: PMC5560134 DOI: 10.7150/jca.20480] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 06/04/2017] [Indexed: 12/26/2022] Open
Abstract
Osteopontin (OPN), a multifunctional protein, has emerged as a potentially valuable biomarker for diagnosing and treating cancers. Recent research focuses on its involvement in tumor biology including the cell proliferation, survival, angiogenesis, invasion, and metastasis. Understanding the molecular mechanisms and pharmacological effects of OPN in cancer development could lead to new targets for improving cancer diagnosis and treatment. This review explains how the structurally conserved domains of OPN are associated with OPN signaling mediators and CD44, and how the conserved OPN domains determine biological functions. The authors have reviewed representative works of OPN expression in breast cancer and colorectal cancer to elucidate the relationship between OPN and cancer/tumor biology. It has also been shown that the prognostic sensitivity in non-small cell lung cancer, hepatocellular carcinoma, gastric cancer, and ovarian cancer improved compared to the individual marker when OPN was analyzed in conjunction with other markers. The therapeutic approaches based on OPN inhibitors are discussed to illustrate recent research progress. Previous clinical data has indicated that OPN has played a unique role in cancer development, but further investigation is required to understand the underlying mechanism. More clinical trials are also required to examine the applicability and efficacy of OPN inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Ran Wei
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Janet Pik Ching Wong
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| |
Collapse
|
50
|
Hao C, Cui Y, Owen S, Li W, Cheng S, Jiang WG. Human osteopontin: Potential clinical applications in cancer (Review). Int J Mol Med 2017; 39:1327-1337. [PMID: 28440483 PMCID: PMC5428945 DOI: 10.3892/ijmm.2017.2964] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022] Open
Abstract
Human osteopontin (OPN) is a glycosylated phosphoprotein which is expressed in a variety of tissues in the body. In recent years, accumulating evidence has indicated that the aberrant expression of OPN is closely associated with tumourigensis, progression and most prominently with metastasis in several tumour types. In this review, we present the current knowledge on the expression profiles of OPN and its main splice variants in human cancers, as well as the potential implications in patient outcome. We also discuss its putative clinical application as a cancer biomarker and as a therapeutic target.
Collapse
Affiliation(s)
- Chengcheng Hao
- Department of Biochemistry and Molecular Biology
- Beijing Key Laboratory of Cancer and Metastasis Research, Capital Medical University, Beijing 100069, P.R. China
| | - Yuxin Cui
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Sionen Owen
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Wenbin Li
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Shan Cheng
- Department of Biochemistry and Molecular Biology
- Beijing Key Laboratory of Cancer and Metastasis Research, Capital Medical University, Beijing 100069, P.R. China
| | - Wen G. Jiang
- Correspondence to: Professor Wen G. Jiang, Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Henry Wellcome Building, Heath Park Way, Cardiff CF14 4XN, UK, E-mail:
| |
Collapse
|