1
|
Xu J, Jia Q, Du N, Liang J, Wang H, Chai J, Yang Y, Xu T, Ma J, Ji P, Ma Y, Liu B, Yang W, Ma J, Yang Y, Li M. STC1 promotes esophageal squamous cell carcinoma progression and is associated with poor prognosis. Pathol Res Pract 2025; 265:155744. [PMID: 39612809 DOI: 10.1016/j.prp.2024.155744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a malignant tumor that poses a significant threat to human health. Patients are often diagnosed at advanced stages of the disease, resulting in poor clinical outcomes and a short survival period. Recent advances have revealed that ESCC tumors exhibit distinct molecular biological characteristics. Our study investigated the expression and biological function of Stanniocalcin-1 (STC1) in ESCC. METHODS We collected paraffin-embedded tumor tissues from 127 patients with ESCC at Xijing Hospital, as well as fresh tissue specimens from 21 patients who underwent radical resection of ESCC, including both tumor and adjacent normal tissues. The expression levels of STC1 in ESCC tissues and cells were assessed using immunohistochemistry (IHC) and Western blot analysis. We employed Kaplan-Meier survival analysis to explore the impact of STC1 expression on the prognosis of ESCC patients. Additionally, we evaluated the effect of STC1 expression on the malignant behavior of ESCC cells through both in vivo and in vitro experiments. RESULTS Compared to normal esophageal tissue, STC1 is overexpressed in ESCC tissue. Univariate and multivariate analyses of clinical data indicated that patients with STC1 overexpression had a poor prognosis (P = 0.009 and P = 0.015). Both cell experiments and xenograft models demonstrated that the upregulation of STC1 may promote the malignant behavior of ESCC, and conversely, its downregulation may inhibit such behavior. CONCLUSION The overexpression of STC1 enhances the migration, invasion and proliferation of ESCC cells, and is significantly associated with poor prognosis in ESCC patients. Therefore, STC1 may serve as a promising prognostic factor and could also be a potential target for ESCC-specific therapy.
Collapse
Affiliation(s)
- Junpeng Xu
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China
| | - Na Du
- No.2 Department of Medical Care, No.971 Hospital of the PLA Navy, Qingdao, China
| | - Jiayi Liang
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Huanhuan Wang
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Jia Chai
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanru Yang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianwu Ma
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Puzhong Ji
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Yingchun Ma
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Bin Liu
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China
| | - Weiyi Yang
- Department of Neurology, Xi'an Daxing Hospital, Xi'an, China
| | - Jing Ma
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Yanli Yang
- Department of Pathology, The 940th Hospital of the Joint Logistics Support Force, Lanzhou, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
2
|
Wang L, Wang J, Qiang W, Ge W. Stanniocalcin-1 in tumor immunity: acts via macrophages. Front Immunol 2024; 15:1510182. [PMID: 39654892 PMCID: PMC11625730 DOI: 10.3389/fimmu.2024.1510182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
Tumor immune escape has become a research hotspot in the field of cancer immunotherapy. Tumor-associated macrophages (TAMs) are the key component of tumor microenvironment, which play a pivotal role in tumor immune escape by regulating the immunity checkpoints, inhibiting the activity of T lymphocytes and natural killer (NK) cells, and modulating proportion of different T cells. Stanniocalcin-1(STC1)is ubiquitously expressed in human body, which is proven to involve with tumor progression and clinical prognosis. Recently, STC1 is implicated in tumor microenvironment as a phagocytosis checkpoint, as well as regulates the immunity via macrophages. In the review, we discussed the role of STC1 and TAMs in tumor immunity and their crosstalk, hoping to provide references for the research of STC1 in tumor immunotherapy.
Collapse
Affiliation(s)
- Lele Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jianjun Wang
- Department of Pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Weijie Qiang
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Weihong Ge
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Xu C, Li S, Chen H, Chi L, Wang X, He M, Wang Q, Zhang X, Lin Y, Xue F. Integrative analysis of recurrence related gene signature and STC1 in colorectal cancer proliferation and metastasis. J Cancer 2024; 15:6724-6739. [PMID: 39668832 PMCID: PMC11632977 DOI: 10.7150/jca.102605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/13/2024] [Indexed: 12/14/2024] Open
Abstract
Colorectal cancer remains a formidable global health challenge, characterized by high recurrence rates and poor prognosis. This study introduces a novel Recurrence Related Gene Signature (RRGS), designed to predict therapy response and enhance prognostic accuracy in colorectal cancer. Through analysis of the GSE17536 cohort, we identified 79 differentially expressed genes (DEGs) between recurrent and non-recurrent cases, comprising 54 upregulated and 25 downregulated genes. Pathway analysis revealed that upregulated genes were enriched in cancer progression-related pathways, while downregulated genes were associated with immune-related processes. Leveraging these findings, we developed the RRGS using LASSO regression, resulting in an innovative 11-gene model with robust diagnostic and prognostic capabilities. Notably, the RRGS demonstrated significant predictive value for both overall survival and disease-free survival across multiple datasets, with higher RRGS scores correlating with advanced tumor stages and poorer outcomes, particularly in post-chemotherapy patients. This predictive power highlights the RRGS's potential in guiding personalized treatment strategies. Furthermore, we identified STC1 as a critical component of the RRGS, playing a significant role in tumor progression and immune evasion. Through rigorous in vitro and in vivo experiments we confirmed that STC1 knockdown substantially reduced cell proliferation and metastasis, emphasizing its potential as a therapeutic target. This comprehensive study not only elucidates the molecular mechanisms driving colorectal cancer recurrence but also introduces a powerful tool for enhancing prognostic accuracy and personalizing therapeutic interventions.
Collapse
Affiliation(s)
- Chao Xu
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| | - ShuYuan Li
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| | - HongYuan Chen
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| | - LiangJie Chi
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| | - XiangYu Wang
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| | - Muzhen He
- Department of Radiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| | - Qingshui Wang
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiuli Zhang
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - FangQin Xue
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, No. 134 Dongjie, Fuzhou, China
| |
Collapse
|
4
|
Li Q, Xu Z, Gong Q, Shen X. Identification and Validation of STC1 Act as a Biomarker for High-Altitude Diseases and Its Pan-Cancer Analysis. Int J Mol Sci 2024; 25:9085. [PMID: 39201771 PMCID: PMC11354978 DOI: 10.3390/ijms25169085] [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: 07/26/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
High-altitude diseases, including acute mountain sickness (AMS), high-altitude cerebral edema (HACE), and high-altitude pulmonary edema (HAPE), are closely related to an individual's ability to adapt to hypoxic environments. However, specific research in this field is relatively limited, and further biomarker research and clinical trials are needed to clarify the exact role and potential therapeutic applications of key genes in high-altitude diseases. This study focuses on the role of the STC1 gene in high-altitude diseases and explores its expression patterns in different types of cancer. By using gene expression data analysis and functional experiments, we identified STC1 as a key gene affecting the development of altitude sickness. In addition, we also conducted expression and mutation analysis on STC1 in various cancer samples and found significant differences in the expression of this gene in 13 types of malignant tumors, which is associated with the hypoxic state in the tumor microenvironment. In addition, STC1 is significantly associated with patient prognosis and influences tumor immunity by mediating six types of immune cells (CD8+T cells, CD4+T cells, neutrophils, macrophages, monocytes, and B cells) in the tumor microenvironment. The expression and diagnostic value of STC1 were confirmed through GEO datasets and qPCR testing, indicating consistency with the results of bioinformatics analysis. These results indicate that STC1 is not only an important factor in the adaptive response to high-altitude diseases but may also play a role in the adaptation of cancer to low-oxygen environments. Our research provides a new perspective and potential targets for the discovery of biomarkers for high-altitude diseases and cancer treatment.
Collapse
Affiliation(s)
- Qiong Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210003, China; (Q.L.); (Z.X.); (Q.G.)
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing 210003, China
| | - Zhichao Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210003, China; (Q.L.); (Z.X.); (Q.G.)
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing 210003, China
| | - Qianhui Gong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210003, China; (Q.L.); (Z.X.); (Q.G.)
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing 210003, China
| | - Xiaobing Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210003, China; (Q.L.); (Z.X.); (Q.G.)
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing 210003, China
| |
Collapse
|
5
|
Sun D, Gong L, Wang X, Chen S, Yi J, Liu X. Pro-inflammatory Cytokines Promote the Occurrence and Development of Colitis-associated Colorectal Cancer by Inhibiting miR-615-5p. Inflamm Bowel Dis 2023; 29:1854-1864. [PMID: 37300504 DOI: 10.1093/ibd/izad105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Indexed: 06/12/2023]
Abstract
BACKGROUND Patients with ulcerative colitis (UC) may be prone to colitis-associated colorectal cancer (CAC), but there is still a poor understanding of the underlying mechanism so far. This study intended to clarify the role of pro-inflammatory cytokines and miR-615-5p in this process. METHODS This experiment first detected miR-615-5p expressions in paraffin-embedded sections of colonic tissues from patients with UC and CAC. Then, we investigated the mechanism through which pro-inflammatory cytokines affected miR-615-5p. Furthermore, in vivo and in vitro tests were performed to identify how miR-615-5p affected colorectal cancer (CRC). Dual-luciferase reporter assay was then employed to identify the targeting relationship between miR-615-5p and stanniocalcin-1 (STC1). RESULTS The miR-615-5p was lowly expressed in both cancerous and noncancerous colonic tissues of patients with CAC. Pro-inflammatory cytokines downregulated miR-615-5p expression. Overexpression of miR-615-5p reduced the proliferation and migration of CRC cells and had a certain therapeutic effect on in human CRC xenograft mice. Stanniocalcin-1 was identified to be a target gene of miR-615-5p and was involved in the effect of miR-615-5p on CRC. CONCLUSIONS During the progression from UC to CAC, pro-inflammatory cytokines downregulate miR-615-5p, which may induce the upregulation of STC1, and promote the occurrence and development of tumors. These findings offer new insights into the mechanism of CAC and may indicate novel tumor markers or therapeutic targets.
Collapse
Affiliation(s)
- Danping Sun
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lingqi Gong
- Department of Gastroenterology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Xiaotong Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuijiao Chen
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Jun Yi
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
6
|
Bajwa P, Kordylewicz K, Bilecz A, Lastra RR, Wroblewski K, Rinkevich Y, Lengyel E, Kenny HA. Cancer-associated mesothelial cell-derived ANGPTL4 and STC1 promote the early steps of ovarian cancer metastasis. JCI Insight 2023; 8:e163019. [PMID: 36795484 PMCID: PMC10070116 DOI: 10.1172/jci.insight.163019] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Ovarian cancer (OvCa) preferentially metastasizes in association with mesothelial cell-lined surfaces. We sought to determine if mesothelial cells are required for OvCa metastasis and detect alterations in mesothelial cell gene expression and cytokine secretion upon interaction with OvCa cells. Using omental samples from patients with high-grade serous OvCa and mouse models with Wt1-driven GFP-expressing mesothelial cells, we validated the intratumoral localization of mesothelial cells during human and mouse OvCa omental metastasis. Removing mesothelial cells ex vivo from human and mouse omenta or in vivo using diphtheria toxin-mediated ablation in Msln-Cre mice significantly inhibited OvCa cell adhesion and colonization. Human ascites induced angiopoietin-like 4 (ANGPTL4) and stanniocalcin 1 (STC1) expression and secretion by mesothelial cells. Inhibition of STC1 or ANGPTL4 via RNAi obstructed OvCa cell-induced mesothelial cell to mesenchymal transition while inhibition of ANGPTL4 alone obstructed OvCa cell-induced mesothelial cell migration and glycolysis. Inhibition of mesothelial cell ANGPTL4 secretion via RNAi prevented mesothelial cell-induced monocyte migration, endothelial cell vessel formation, and OvCa cell adhesion, migration, and proliferation. In contrast, inhibition of mesothelial cell STC1 secretion via RNAi prevented mesothelial cell-induced endothelial cell vessel formation and OvCa cell adhesion, migration, proliferation, and invasion. Additionally, blocking ANPTL4 function with Abs reduced the ex vivo colonization of 3 different OvCa cell lines on human omental tissue explants and in vivo colonization of ID8p53-/-Brca2-/- cells on mouse omenta. These findings indicate that mesothelial cells are important to the initial stages of OvCa metastasis and that the crosstalk between mesothelial cells and the tumor microenvironment promotes OvCa metastasis through the secretion of ANGPTL4.
Collapse
Affiliation(s)
- Preety Bajwa
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology
| | | | - Agnes Bilecz
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology
- Department of Pathology, and
| | | | - Kristen Wroblewski
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology
| | - Hilary A. Kenny
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology
| |
Collapse
|
7
|
Sengun S, Korkmaz H, Ciris M, Yüceer RO, Boyluboy SM, Kiran M. Diagnostic and prognostic value of Stanniocalcin 1 expression in papillary thyroid cancer. Endocrine 2022; 78:95-103. [PMID: 35788886 DOI: 10.1007/s12020-022-03126-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE To evaluate the potential role of immunohistochemical changes in stanniocalcin 1 (STC1) and stanniocalcin 2 (STC2) expressions in papillary thyroid cancer (PTC) tissues in the disease's diagnosis and to investigate their relationship with classical clinicopathological prognostic factors. METHODS The study included 100 patients with PTC. Normal thyroid tissue adjacent to the tumor was taken as the control group. Clinicopathological prognostic features at the time of diagnosis of patients were recorded. STC1 and STC2 expressions of tumor tissue and adjacent normal tissue were determined immunohistochemically. RESULTS The sensitivity of STC1 in the diagnosis of PTC was 93%, the specificity was 94%, positive predictive value (PPV) 93.9%, and negative predictive value (NPV) 93.1%. It was determined that the STC1 staining score in tumor tissue was positively correlated with the disease TNM stage score (r = 0.259, p = 0.009) and the increase in STC1 staining score were independent risk factors that increased the risk of lymph node metastasis (R2 = 0.398, p < 0.001). While 21% of the tumor tissues were stained with STC2, none of the normal thyroid tissues adjacent to the tumor tissue showed any staining with STC2. No correlation was found between STC2 immunohistochemical staining of tumor tissue and clinicopathological risk factors for the disease. CONCLUSION Increased STC1 expression in thyroid lesions may be helpful in diagnosing PTC. In addition, since increased STC1 expression in PTC tissues is associated with the risk of lymph node metastasis, it may be an efficient marker for predicting the prognosis of the disease.
Collapse
Affiliation(s)
- Sevinç Sengun
- Department of Internal Medicine, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Hakan Korkmaz
- Department of Internal Medicine, Division of Endocrinology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey.
| | - Metin Ciris
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Ramazan Oguz Yüceer
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Serife Mehtap Boyluboy
- Department of Internal Medicine, Division of Endocrinology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| | - Mehmet Kiran
- Department of Pathology, Faculty of Medicine, Suleyman Demirel University, 32260, Cunur, Isparta, Turkey
| |
Collapse
|
8
|
Identification and validation of a prognostic signature related to hypoxic tumor microenvironment in cervical cancer. PLoS One 2022; 17:e0269462. [PMID: 35657977 PMCID: PMC9165826 DOI: 10.1371/journal.pone.0269462] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 05/17/2022] [Indexed: 12/24/2022] Open
Abstract
Background Hypoxia is a common microenvironment condition in most malignant tumors and has been shown to be associated with adverse outcomes of cervical cancer patients. In this study, we investigated the effects of hypoxia-related genes on tumor progress to characterize the tumor hypoxic microenvironment. Methods We retrieved a set of hypoxia-related genes from the Molecular Signatures Database and evaluated their prognostic value for cervical cancer. A hypoxia-based prognostic signature for cervical cancer was then developed and validated using tumor samples from two independent cohorts (TCGA-CESC and CGCI-HTMCP-CC cohorts). Finally, we validated the hypoxia prediction of ccHPS score in eight human cervical cancer cell lines treated with the hypoxic and normoxic conditions, and 286 tumor samples with hypoxic category (more or less) from Gene Expression Omnibus (GEO) database with accession GSE72723. Results A risk signature model containing nine hypoxia-related genes was developed and validated in cervical cancer. Further analysis showed that this risk model could be an independent prognosis factor of cervical cancer, which reflects the condition of the hypoxic tumor microenvironment and its remodeling of cell metabolism and tumor immunity. Furthermore, a nomogram integrating the novel risk model and lymphovascular invasion status was developed, accurately predicting the 1-, 3- and 5-year prognosis with AUC values of 0.928, 0.916 and 0.831, respectively. These findings provided a better understanding of the hypoxic tumor microenvironment in cervical cancer and insights into potential new therapeutic strategies in improving cancer therapy.
Collapse
|
9
|
Li R, Liu R, Wu S, Zheng S, Ye L, Shao Y. Prognostic value of STC1 in solid tumors: a meta-analysis. Biomark Med 2022; 16:253-263. [PMID: 35176895 DOI: 10.2217/bmm-2021-0835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: The prognostic value of STC1 has been evaluated in solid tumors. However, the results remain controversial. Materials & methods: Relevant studies published up to 27 February 2021 were identified by a comprehensive search of the PubMed, EMBASE and Web of Science databases. Hazard ratios (HRs) and odds ratios with 95% CIs were applied to explore the association between STC1 and survival outcome and clinical characteristics. Results: Sixteen articles involving 2942 participants were included in this meta-analysis. The pooled analysis showed that high STC1 expression was significantly associated with worse overall survival (HR: 1.91; 95% CI: 1.63-2.24) and disease-free survival/progression-free survival/relapse-free survival (HR: 2.01; 95% CI: 1.34-3.02). Conclusion: STC1 may be an effective prognostic marker in solid tumors.
Collapse
Affiliation(s)
- Rongqi Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.,Department of Hepatobiliary Surgery, Foshan hospital of Traditional Chinese Medical University, Foshan, Guangdong, 528000, China
| | - Rongqiang Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510220, China
| | - Shinan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shiyang Zheng
- Department of breast surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lei Ye
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| |
Collapse
|
10
|
Khatun M, Urpilainen E, Ahtikoski A, Arffman RK, Pasanen A, Puistola U, Tapanainen JS, Andersson LC, Butzow R, Loukovaara M, Piltonen TT. Low Expression of Stanniocalcin 1 (STC-1) Protein Is Associated With Poor Clinicopathologic Features of Endometrial Cancer. Pathol Oncol Res 2021; 27:1609936. [PMID: 34650342 PMCID: PMC8505533 DOI: 10.3389/pore.2021.1609936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022]
Abstract
Stanniocalcin-1 (STC-1) is a glycoprotein hormone involved in diverse biological processes, including regulation of calcium phosphate homeostasis, cell proliferation, apoptosis, inflammation, oxidative stress responses, and cancer development. The role of STC-1 in endometrial cancer (EC) is yet to be elucidated. In this study, we investigated the protein expression pattern of STC-1 in a tissue microarray (TMA) cohort of hysterectomy specimens from 832 patients with EC. We then evaluated the prognostic value of STC-1 expression regarding the clinicopathologic features and patients survival over a period of 140 months. Our results revealed that in EC tissue samples, STC-1 is mainly localized in the endometrial epithelium, although some expression was also observed in the stroma. Decreased STC-1 expression was associated with factors relating to a worse prognosis, such as grade 3 endometrioid tumors (p = 0.030), deep myometrial invasion (p = 0.003), lymphovascular space invasion (p = 0.050), and large tumor size (p = 0.001). Moreover, STC-1 expression was decreased in tumors obtained from obese women (p = 0.014) and in women with diabetes mellitus type 2 (DMT2; p = 0.001). Interestingly, the data also showed an association between DNA mismatch repair (MMR) deficiency and weak STC-1 expression, specifically in the endometrial epithelium (p = 0.048). No association was observed between STC-1 expression and disease-specific survival. As STC-1 expression was particularly low in cases with obesity and DMT2 in the TMA cohort, we also evaluated the correlation between metformin use and STC-1 expression in an additional EC cohort that only included women with DMT2 (n = 111). The analysis showed no difference in STC-1 expression in either the epithelium or the stroma in women undergoing metformin therapy compared to metformin non-users. Overall, our data may suggest a favorable role for STC-1 in EC behavior; however, further studies are required to elucidate the detailed mechanism and possible applications to cancer treatment.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Elina Urpilainen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Anne Ahtikoski
- Department of Pathology, Oulu University Hospital, University of Oulu, Oulu, Finland.,Department of Pathology, Turku University Hospital, Turku, Finland
| | - Riikka K Arffman
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Annukka Pasanen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Ulla Puistola
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,Department of Obstetrics and Gynaecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Leif C Andersson
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Ralf Butzow
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mikko Loukovaara
- Department of Obstetrics and Gynaecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Terhi T Piltonen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
11
|
Sun J, Wei X, You J, Yue W, Ouyang J, Ling Z, Hou J. STC1 is a Novel Biomarker Associated with Immune Characteristics and Prognosis of Bladder Cancer. Int J Gen Med 2021; 14:5505-5516. [PMID: 34539184 PMCID: PMC8445105 DOI: 10.2147/ijgm.s329723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Background Stanniocalcin-1 (STC1) is a well-studied oncogene that promotes different types of cancer progression. However, the expression status of STC1, the values of STC1 on prognosis, and its immune characteristic in bladder cancer (BLCA) have not been well examined. Methods The expression of STC1 and its clinicopathological as well as immune characteristics in BLCA samples were firstly identified in The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Immunohistochemistry (IHC) performed on the tissue microarray (TMA) slide was further used to validate the expression of STC1 and its relationship with immune features in 16 non-muscle invasive bladder cancer (NMIBC) samples and 42 muscle invasive bladder cancer (MIBC) samples. Results The expression of STC1 was upregulated in higher stage BLCA. High STC1 expression also predicted poor prognosis in BLCA. Subsequently, the TMA validated the expression and prognostic value of STC1 in BLCA. Bioinformatics analysis demonstrated that STC1 and common immune checkpoints as well as immune markers of various immune cells were positively correlated in TCGA. In addition, IHC data from the TMA further validated that tumor cells with higher STC1 level tended to express higher PDL1 as well as increased infiltration of CD3+ T cells. Conclusion To our knowledge, this is the first comprehensive study that investigates the clinical and immune characteristics of STC1 in BLCA. It may provide new insight into the function of STC1 in regulating tumor immune microenvironment. Further studies are warranted to uncover the potential mechanisms that mediate STC1 expression and tumor immunity in BLCA.
Collapse
Affiliation(s)
- Jiale Sun
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jiawei You
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Wenchang Yue
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Ouyang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhixin Ling
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
12
|
Tan GF, Goh S, Lim AH, Liu W, Lee JY, Rajasegaran V, Sam XX, Tay TKY, Selvarajan S, Ng CCY, Teh BT, Chan JY. Bizarre giant cells in human angiosarcoma exhibit chemoresistance and contribute to poor survival outcomes. Cancer Sci 2020; 112:397-409. [PMID: 33164299 PMCID: PMC7780052 DOI: 10.1111/cas.14726] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 11/28/2022] Open
Abstract
Giant cells (GC) are a poorly understood subset of tumor cells that have been increasingly recognized as a potential contributor to tumor heterogeneity and treatment resistance. We aimed to characterize the biological and clinical significance of GC in angiosarcoma, an aggressive rare cancer of endothelial origin. Archival angiosarcoma samples were examined for the presence of GC and compared with clinicopathological as well as NanoString gene expression data. GC were examined in angiosarcoma cell lines MOLAS and ISOHAS using conventional and electron microscopy, single cell whole genome profiling, and other assays. In the cell lines, GC represented a rare population of mitotically active, non–senescent CD31+ cells, and shared similar genomic profiles with regular‐sized cells, consistent with a malignant endothelial phenotype. GC remained viable and persisted in culture following exposure to paclitaxel and doxorubicin. In patient samples, GC were present in 24 of 58 (41.4%) cases. GC was correlated with poorer responses to chemotherapy (25.0% vs 73.3%, P = 0.0213) and independently contributed to worse overall survival outcomes (hazard ratio 2.20, 95% confidence interval 1.17‐4.15, P = 0.0142). NanoString profiling revealed overexpression of genes, including COL11A1, STC1, and ERO1A, accompanied by upregulation of immune‐related metabolic stress and metastasis/matrix remodeling pathways in GC‐containing tumors. In conclusion, GC may contribute to chemoresistance and poor prognosis in angiosarcoma.
Collapse
Affiliation(s)
- Grace Fangmin Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore City, Singapore
| | - Shane Goh
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore City, Singapore
| | - Abner Herbert Lim
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore City, Singapore
| | - Wei Liu
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore City, Singapore
| | - Jing Yi Lee
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore City, Singapore
| | - Vikneswari Rajasegaran
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore City, Singapore
| | - Xin Xiu Sam
- Department of Anatomical Pathology, Singapore General Hospital, Singapore City, Singapore
| | - Timothy Kwang Yong Tay
- Department of Anatomical Pathology, Singapore General Hospital, Singapore City, Singapore
| | | | - Cedric Chuan-Young Ng
- Integrated Genomics Platform, National Cancer Centre Singapore, Singapore City, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore City, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore City, Singapore.,Institute of Molecular and Cell Biology, Singapore City, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore City, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore City, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore City, Singapore.,Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore City, Singapore
| |
Collapse
|
13
|
Luo W, Chen D, Wang H, Hu J. Stanniocalcin 1 is a prognostic biomarker in glioma. Oncol Lett 2020; 20:2248-2256. [PMID: 32782542 PMCID: PMC7400771 DOI: 10.3892/ol.2020.11792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 04/01/2020] [Indexed: 12/25/2022] Open
Abstract
Malignant gliomas are the most common type of primary malignancy of the central nervous system with a poor prognosis. Stanniocalcin 1 (STC1) is closely associated with tumor genesis and development. However, its role in the development and progression of glioma is poorly understood. In silico analysis, The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), Rembrandt and GSE16011 datasets were used to assess the expression levels of STC1 in non-tumor brain tissues and gliomas. Moreover, reverse transcription-quantitative PCR and immunohistochemistry were used to detect STC1 expression in tumor tissues collected in the Department of Neurosurgery of Shenzhen People's Hospital (Shenzhen, China). The association between STC1 expression and different molecular pathological features was analyzed in four public datasets, as well as via Kaplan-Meier analysis. Furthermore, normalized mRNA expression in TCGA was used to perform Gene Ontology analysis. It was revealed that STC1 expression was significantly elevated in glioma tissues compared with the non-tumor brain tissues, both in silico analysis and via cohort validation. According to TCGA, CGGA, Rembrandt and GSE16011 datasets, it was identified that STC1 expression was increased in high grade glioma compared with low grade glioma. In addition, the results indicated STC1 expression was enriched in the isocitrate dehydrogenase (IDH) wild-type and mesenchymal subtype in TCGA, GSE16011 and Rembrandt datasets. Moreover, it was demonstrated that patients with higher STC1 expression exhibited shorter overall survival times compared with those with lower STC1 expression using Kaplan-Meier analysis, according to both the public datasets and validation cohort. Furthermore, the results of the Gene Ontology analysis demonstrated that STC1 was primarily involved in the reorganization of extracellular matrix and was significantly correlated with invasive-related proteins. Therefore, the present results indicate that STC1 was upregulated in glioma tissues and may represent a prognostic biomarker in patients with glioma.
Collapse
Affiliation(s)
- Weijian Luo
- Department of Neurosurgery, Shenzhen People's Hospital, Second Clinical Medical College of Ji'nan University, Shenzhen, Guangdong 518020, P.R. China
| | - Dong Chen
- Department of Neurosurgery, Shenzhen People's Hospital, Second Clinical Medical College of Ji'nan University, Shenzhen, Guangdong 518020, P.R. China
| | - Hao Wang
- Department of Neurosurgery, Shenzhen People's Hospital, Second Clinical Medical College of Ji'nan University, Shenzhen, Guangdong 518020, P.R. China
| | - Jiliang Hu
- Department of Neurosurgery, Shenzhen People's Hospital, Second Clinical Medical College of Ji'nan University, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
14
|
Kamata T, So TY, Ahmed Q, Giblett S, Patel B, Luo J, Reddel R, Pritchard C. Fibroblast-Derived STC-1 Modulates Tumor-Associated Macrophages and Lung Adenocarcinoma Development. Cell Rep 2020; 31:107802. [PMID: 32579928 PMCID: PMC7326292 DOI: 10.1016/j.celrep.2020.107802] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/20/2020] [Accepted: 06/01/2020] [Indexed: 11/25/2022] Open
Abstract
The tumor microenvironment (TME) consists of different cell types, including tumor-associated macrophages (TAMs) and tumor-associated fibroblasts (TAFs). How these cells interact and contribute to lung carcinogenesis remains elusive. Using G12DKRAS- and V600EBRAF-driven mouse lung models, we identify the pleiotropic glycoprotein stanniocalcin-1 (STC1) as a regulator of TAM-TAF interactions. STC1 is secreted by TAFs and suppresses TAM differentiation, at least in part, by sequestering the binding of GRP94, an autocrine macrophage-differentiation-inducing factor, to its cognate scavenger receptors. The accumulation of mature TAMs in the Stc1-deficient lung leads to enhanced secretion of TGF-β1 and, thus, TAF accumulation in the TME. Consistent with the mouse data, in human lung adenocarcinoma, STC1 expression is restricted to myofibroblasts, and a significant increase of naive macrophages is detected in STC1-high compared with STC1-low cases. This work increases our understanding of lung adenocarcinoma development and suggests new approaches for therapeutic targeting of the TME.
Collapse
Affiliation(s)
- Tamihiro Kamata
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK.
| | - Tsz Y So
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Qasim Ahmed
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Susan Giblett
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Bipin Patel
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Jinli Luo
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Roger Reddel
- Cancer Research Unit, Children's Medical Research Institute, University of Sydney, Westmead, NSW, Australia
| | - Catrin Pritchard
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK.
| |
Collapse
|
15
|
Zhao F, Yang G, Feng M, Cao Z, Liu Y, Qiu J, You L, Zheng L, Zhang T, Zhao Y. Expression, function and clinical application of stanniocalcin-1 in cancer. J Cell Mol Med 2020; 24:7686-7696. [PMID: 32468698 PMCID: PMC7348177 DOI: 10.1111/jcmm.15348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein stanniocalcin-1 functions as a regulatory endocrine hormone that maintains the balance of calcium and phosphorus in bony fish and as a paracrine/autocrine factor involved in many physiological/pathological processes in humans, including carcinogenesis. In this review, we provide an overview of (a) the possible mechanisms through which STC1 affects the malignant properties of cancer, (b) transcriptional and post-transcriptional regulation pathways of STC1 and (c) the potential clinical relevance of STC1 as a cancer biomarker and even a therapeutic target in the future. Exploring the role of STC1 in cancer development may provide a better understanding of the tumorigenesis process in humans and may facilitate finding an effective therapeutic method against cancer.
Collapse
Affiliation(s)
- Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Ezure T, Amano S. Stanniocalcin-1 mediates negative regulatory action of epidermal layer on expression of matrix-related genes in dermal fibroblasts. Biofactors 2019; 45:944-949. [PMID: 31348577 PMCID: PMC6916204 DOI: 10.1002/biof.1547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/09/2019] [Indexed: 11/11/2022]
Abstract
Dermal-epidermal interaction plays a role in many pathophysiological processes, such as tumor invasion and psoriasis, as well as wound healing, and is mediated at least in part by secretory factors. In this study, we investigated the factor(s) involved. We found that stanniocalcin-1 (STC1), a cytokine, is expressed at the basal layer of epidermis. Knockdown of STC1 with siRNA in HaCaT cells decreased matrix metalloproteinase 1 (MMP1) expression, suggesting that STC1 serves as an autocrine factor, maintaining MMP1 mRNA expression in the epidermal layer. In dermal fibroblasts, STC1 increased MMP1 mRNA expression and decreased collagen1A1 and elastin mRNA expression. These actions were inhibited by SP600125, a jun kinase (JNK) inhibitor. Nuclear translocation of AP-1, a downstream signal of JNK, was implicated in the actions of STC1. In a coculture system of HaCaT cells and fibroblasts, used as a model of dermal-epidermal interaction, knockdown of STC1 in HaCaT cells with siRNA reduced the negative effects (i.e., induction of MMP1 and decrease of collagen1A1 and elastin) of STC1 on fibroblasts. These results suggest that STC1 secreted from the epidermal layer is a mediator of dermal-epidermal interaction.
Collapse
Affiliation(s)
| | - Satoshi Amano
- Shiseido Global Innovation Research CenterYokohamaJapan
| |
Collapse
|
17
|
Chen F, Zhang Z, Pu F. Role of stanniocalcin-1 in breast cancer. Oncol Lett 2019; 18:3946-3953. [PMID: 31579413 PMCID: PMC6757304 DOI: 10.3892/ol.2019.10777] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 07/16/2019] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is a highly heterogeneous disease consisting of five disease subtypes with distinct histological characteristics, clinical behaviors and prognostic features. Stanniocalcin-1 (STC1) is a secreted glycoprotein hormone that has been demonstrated to regulate calcium and phosphate homeostasis. Mammalian STC1 is expressed in various tissues and is implicated in multiple physiological and pathophysiological processes. In addition, growing evidence has suggested that STC1 serves an oncogenic role in a number of different types of tumor. However, the role of STC1 in breast cancer is complex, considering that some studies have shown that it exerts an oncogenic role, whereas other studies have demonstrated the opposite. The aim of the present review article is to evaluate the currently available data on mammalian STC1 and discuss its potential roles in each subtype of breast cancer.
Collapse
Affiliation(s)
- Fengxia Chen
- Department of Medical Oncology, General Hospital of The Yangtze River Shipping, Wuhan, Hubei 430010, P.R. China.,Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Feifei Pu
- Department of Orthopedics, Wuhan No. 1 Hospital, Wuhan Integrated Traditional Chinese Medicine and Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
18
|
Chan KKS, Leung CON, Wong CCL, Ho DWH, Chok KSH, Lai CL, Ng IOL, Lo RCL. Secretory Stanniocalcin 1 promotes metastasis of hepatocellular carcinoma through activation of JNK signaling pathway. Cancer Lett 2017; 403:330-338. [PMID: 28688970 DOI: 10.1016/j.canlet.2017.06.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/19/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022]
Abstract
The hypoxic microenvironment is well-characterized in hepatocellular carcinoma (HCC). Delineation of hypoxia-responsive events is an integral part to understand the pathogenesis of HCC. We studied the functional role and clinical relevance of Stanniocalcin 1 (STC1), a hypoxia-induced molecular target, in HCC. In our clinical cohort, STC1 transcript was up-regulated in HCC tumor tissues. Moreover, STC1 protein was detected in the sera of HCC patients. A higher serum STC1 level was correlated with larger tumor size and poorer 5-year disease-free survival. Functionally, recombinant STC1 protein (rhSTC1) promoted cell migration and cell invasion in vitro; and the effect was abolished by co-treatment of anti-STC1 neutralizing antibody. By in vivo mouse model, silencing of STC1 in HCC cells downregulated secretory STC1 level and suppressed lung metastasis. Furthermore, we found that rhSTC1 activated the JNK pathway, as evidenced by altered expression of the key molecular targets pJNK and p-c-Jun. The functional effects conferred by rhSTC1 were abrogated by co-treatment of JNK inhibitor. In summary, secretory STC1 enhances metastatic potential of HCC via JNK signaling. It potentially serves as a prognostic serum biomarker and a therapeutic target for HCC.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/secondary
- Cell Movement/drug effects
- Disease-Free Survival
- Gene Expression Regulation, Neoplastic
- Glycoproteins/blood
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Humans
- JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors
- JNK Mitogen-Activated Protein Kinases/metabolism
- Kaplan-Meier Estimate
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Lung Neoplasms/enzymology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mice, Inbred BALB C
- Mice, Nude
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- RNA Interference
- Signal Transduction/drug effects
- Time Factors
- Transfection
- Tumor Burden
- Tumor Hypoxia
- Tumor Microenvironment
- Up-Regulation
Collapse
Affiliation(s)
- Kristy Kwan-Shuen Chan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Carmen Oi-Ning Leung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Carmen Chak-Lui Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Daniel Wai-Hung Ho
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kenneth Siu-Ho Chok
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ching-Lung Lai
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Regina Cheuk-Lam Lo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
19
|
Wang C, Wang J, Chen Z, Gao Y, He J. Immunohistochemical prognostic markers of esophageal squamous cell carcinoma: a systematic review. CHINESE JOURNAL OF CANCER 2017; 36:65. [PMID: 28818096 PMCID: PMC5561640 DOI: 10.1186/s40880-017-0232-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 04/17/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy, with a high incidence and poor prognosis. In the past several decades, hundreds of proteins have been reported to be associated with the prognosis of ESCC, but none has been widely accepted to guide clinical care. This study aimed to identify proteins with great potential for predicting prognosis of ESCC. METHODS We conducted a systematic review on immunohistochemical (IHC) prognostic markers of ESCC according to the 2009 Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) Guidelines. Literature related to IHC prognostic markers of ESCC were searched from PubMed, Embase, Web of Science, and Cochrane Library until January 30th, 2017. The risk of bias of these original studies was evaluated using the Quality in Prognosis Studies (QUIPS) tool. RESULTS We identified 11 emerging IHC markers with reproducible results, including eight markers [epidermal growth factor receptor (EGFR), Cyclin D1, vascular endothelial growth factor (VEGF), Survivin, Podoplanin, Fascin, phosphorylated mammalian target of rapamycin (p-mTOR), and pyruvate kinase M2 (PKM2)] indicating unfavorable prognosis and 3 markers (P27, P16, and E-cadherin) indicating favorable prognosis of ESCC. CONCLUSION Strong evidence supports that these 11 emerging IHC markers or their combinations may be useful in predicting prognosis and aiding personalized therapy decision-making for ESCC patients.
Collapse
Affiliation(s)
- Chunni Wang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Science Building, No.17 Panjiayuan Nanli, Chaoyang District, PO Box 2258, Beijing, 100021 P. R. China
| | - Jingnan Wang
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Science Building, No.17 Panjiayuan Nanli, Chaoyang District, PO Box 2258, Beijing, 100021 P. R. China
| | - Zhaoli Chen
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Science Building, No.17 Panjiayuan Nanli, Chaoyang District, PO Box 2258, Beijing, 100021 P. R. China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Science Building, No.17 Panjiayuan Nanli, Chaoyang District, PO Box 2258, Beijing, 100021 P. R. China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Science Building, No.17 Panjiayuan Nanli, Chaoyang District, PO Box 2258, Beijing, 100021 P. R. China
- Center for Cancer Precision Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021 P. R. China
| |
Collapse
|
20
|
Han J, Jeon M, Shin I, Kim S. Elevated STC-1 augments the invasiveness of triple-negative breast cancer cells through activation of the JNK/c-Jun signaling pathway. Oncol Rep 2016; 36:1764-71. [DOI: 10.3892/or.2016.4977] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/02/2016] [Indexed: 11/06/2022] Open
|
21
|
The oncogenetic role of stanniocalcin 1 in lung adenocarcinoma: a promising serum candidate biomarker for tracking lung adenocarcinoma progression. Tumour Biol 2015; 37:5633-44. [DOI: 10.1007/s13277-015-4431-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022] Open
|
22
|
Yeung BHY, Shek FH, Lee NP, Wong CKC. Stanniocalcin-1 Reduces Tumor Size in Human Hepatocellular Carcinoma. PLoS One 2015; 10:e0139977. [PMID: 26469082 PMCID: PMC4607425 DOI: 10.1371/journal.pone.0139977] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023] Open
Abstract
Growing evidence has revealed high expression levels of stanniocalcin-1 (STC1) in different types of human cancers. Numerous experimental studies using cancer cell lines demonstrated the involvement of STC1 in inflammatory and apoptotic processes; however the role of STC1 in carcinogenesis remains elusive. Hepatocellular carcinoma (HCC) an exemplified model of inflammation-related cancer, represents a paradigm of studying the association between STC1 and tumor development. Therefore, we conducted a statistical analysis on the expression levels of STC1 using clinicopathological data from 216 HCC patients. We found that STC1 was upregulated in the tumor tissues and its expression levels was positively correlated with the levels of interleukin (IL)-6 and IL-8. Intriguingly tumors with greater expression levels of STC1 (tumor/normal ≥ 2) were significantly smaller than the lower level (tumor/normal<2) samples (p = 0.008). A pharmacological approach was implemented to reveal the functional correlation between STC1 and the ILs in the HCC cell-lines. IL-6 and IL-8 treatment of Hep3B cells induced STC1 expression. Lentiviral-based STC1 overexpression in Hep3B and MHCC-97L cells however showed inhibitory action on the pro-migratory effects of IL-6 and IL-8 and reduced size of tumor spheroids. The inhibitory effect of STC1 on tumor growth was confirmed in vivo using the stable STC1-overexpressing 97L cells on a mouse xenograft model. Genetic analysis of the xenografts derived from the STC1-overexpressing 97L cells, showed upregulation of the pro-apoptotic genes interleukin-12 and NOD-like receptor family, pyrin domain-containing 3. Collectively, the anti-inflammatory and pro-apoptotic functions of STC1 were suggested to relate its inhibitory effect on the growth of HCC cells. This study supports the notion that STC1 may be a potential therapeutic target for inflammatory tumors in HCC patients.
Collapse
Affiliation(s)
- Bonnie H. Y. Yeung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Felix H. Shek
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Nikki P. Lee
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chris K. C. Wong
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
- * E-mail:
| |
Collapse
|
23
|
Hayase S, Sasaki Y, Matsubara T, Seo D, Miyakoshi M, Murata T, Ozaki T, Kakudo K, Kumamoto K, Ylaya K, Cheng SY, Thorgeirsson SS, Hewitt SM, Ward JM, Kimura S. Expression of stanniocalcin 1 in thyroid side population cells and thyroid cancer cells. Thyroid 2015; 25:425-36. [PMID: 25647164 PMCID: PMC4390205 DOI: 10.1089/thy.2014.0464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Mouse thyroid side population (SP) cells consist of a minor population of mouse thyroid cells that may have multipotent thyroid stem cell characteristics. However the nature of thyroid SP cells remains elusive, particularly in relation to thyroid cancer. Stanniocalcin (STC) 1 and 2 are secreted glycoproteins known to regulate serum calcium and phosphate homeostasis. In recent years, the relationship of STC1/2 expression to cancer has been described in various tissues. METHOD Microarray analysis was carried out to determine genes up- and down-regulated in thyroid SP cells as compared with non-SP cells. Among genes up-regulated, stanniocalcin 1 (STC1) was chosen for study because of its expression in various thyroid cells by Western blotting and immunohistochemistry. RESULTS Gene expression analysis revealed that genes known to be highly expressed in cancer cells and/or involved in cancer invasion/metastasis were markedly up-regulated in SP cells from both intact as well as partial thyroidectomized thyroids. Among these genes, expression of STC1 was found in five human thyroid carcinoma-derived cell lines as revealed by analysis of mRNA and protein, and its expression was inversely correlated with the differentiation status of the cells. Immunohistochemical analysis demonstrated higher expression of STC1 in the thyroid tumor cell line and thyroid tumor tissues from humans and mice. CONCLUSION These results suggest that SP cells contain a population of cells that express genes also highly expressed in cancer cells including Stc1, which warrants further study on the role of SP cells and/or STC1 expression in thyroid cancer.
Collapse
Affiliation(s)
- Suguru Hayase
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yoshihito Sasaki
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Kuwana East Medical Center, Kuwana, Mie, Japan
| | - Tsutomu Matsubara
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Daekwan Seo
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Bioinformatics Core, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Masaaki Miyakoshi
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Oral Pathobiological Science, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Tsubasa Murata
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Dental and Oral Surgery, Tomakomai City Hospital, Tomakomai, Hokkaido, Japan
| | - Takashi Ozaki
- Department of Pathology, Wakayama Medical University, Wakayama City, Japan
| | - Kennichi Kakudo
- Department of Pathology, Nara Hospital Kinki University Faculty of Medicine, Ikoma, Japan
| | - Kensuke Kumamoto
- Department of Organ Regulatory Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kris Ylaya
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Snorri S. Thorgeirsson
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen M. Hewitt
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Shioko Kimura
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
24
|
Ma X, Gu L, Li H, Gao Y, Li X, Shen D, Gong H, Li S, Niu S, Zhang Y, Fan Y, Huang Q, Lyu X, Zhang X. Hypoxia-induced overexpression of stanniocalcin-1 is associated with the metastasis of early stage clear cell renal cell carcinoma. J Transl Med 2015; 13:56. [PMID: 25740019 PMCID: PMC4337255 DOI: 10.1186/s12967-015-0421-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/28/2015] [Indexed: 11/10/2022] Open
Abstract
Background Although metastasis of clear cell renal cell carcinoma (ccRCC) is predominantly observed in late stage tumors, early stage metastasis of ccRCC can also be found with indefinite molecular mechanism, leading to inappropriate clinical decisions and poor prognosis. Stanniocalcin-1 (STC1) is a glycoprotein hormone involved in calcium/phosphate homeostasis, which regulates various cellular processes in normal development and tumorigenesis. This study aimed to investigate the role and mechanism of regulation of STC1 in the metastasis of early stage ccRCC. Methods STC1 mRNA and protein expression was determined in ccRCC surgical specimens, RCC cell lines, and human kidney tubule epithelial cell line HKC by real-time polymerase chain reaction (RT-PCR) and western blotting. Immunohistochemistry staining (IHC) and immunofluorescence were also used to examine the expression and localization of STC1 in ccRCC tissues and cancer cells. Knockdown and overexpression studies were conducted in vitro in RCC cell lines using small interfering RNAs (siRNA) and lentiviral-mediated gene delivery to evaluate the role of STC1 in cell proliferation, anchorage-dependent and independent growth, cell cycle control, and migration and invasion. Results STC1 mRNA and protein expression were significantly up-regulated in tumors when compared with non-tumor tissues, with the greatest increase in expression observed in metastatic tissues. Clinicopathological analysis revealed that STC1 mRNA expression was associated with Fuhrman tumor grade (P = 0.008) and overall Tumor Node Metastasis (TNM) staging (P = 0.018). STC1 expression was elevated in T1 stage metastatic tumors when compared with localized tumors, and was positively correlated with average tumor diameter. Silencing of STC1 expression by Caki-1 and A498 resulted in the inhibition of cell proliferation, migration, and invasion, meanwhile down-regulation of STC1 impaired epithelial–mesenchymal transition (EMT) of ccRCC cell lines. Overexpression of STC1 in Caki-2 enhanced cell growth and proliferation but not migration and invasion. Further investigation identified hypoxia and HIF-1α as candidate regulators of STC1 expression. Conclusions Our findings demonstrate a role for STC1 in metastasis of early stage ccRCC and suggest that STC1 may be a biomarker of potential value both for the prognosis of this disease and for guiding clinical decisions regarding surgical strategies and adjuvant treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0421-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin Ma
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Liangyou Gu
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Hongzhao Li
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Yu Gao
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Xintao Li
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Donglai Shen
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Huijie Gong
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Shichao Li
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Shaoxi Niu
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Yu Zhang
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Yang Fan
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Qingbo Huang
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Xiangjun Lyu
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| | - Xu Zhang
- Department of Urology/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital/PLA Medical School, Beijing, China.
| |
Collapse
|
25
|
Fang Z, Tian Z, Luo K, Song H, Yi J. Clinical significance of stanniocalcin expression in tissue and serum of gastric cancer patients. Chin J Cancer Res 2014; 26:602-10. [PMID: 25400427 DOI: 10.3978/j.issn.1000-9604.2014.10.08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/25/2014] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Stanniocalcin (STC) has been recognized as a potential biomarker in a variety of cancers. The aim of this study was to examine STC1 and STC2 expression in tumor and serum samples from gastric cancer (GC) patients. METHODS A total of 83 GC patients treated with radical resection were enrolled in this study. Immunohistochemistry was used to detect STC protein expression in paired tumor and adjacent normal tissues. Serum STC levels were determined by enzyme-linked immunosorbent assay (ELISA). The receiver operating characteristics (ROC) curve was constructed to describe diagnostic specificity and sensitivity. RESULTS Both of STC1 and STC2 protein expression were upregulated in GC tissues compared with that in normal ones. Moreover, the high/moderate of STC1 protein was significantly associated with lymph metastasis, clinical stage and adverse 3-year progression-free survival (PFS). In addition, serum STC1 and STC2 expression in GC patients were much higher than that in patients with benign gastric disease, which decreased at postoperative 7-10 days. The sensitivity of serum STC protein also showed superiority over CEA and CA19-9. CONCLUSIONS STC upregulation plays an important role in GC development, and serum STC1 and STC2 might function as promising tumor markers for GC diagnosis and prognosis.
Collapse
Affiliation(s)
- Zheng Fang
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Zhiqiang Tian
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Kunlun Luo
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Haizhu Song
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Jun Yi
- 1 Department of General Surgery, 101 Hospital of People's Liberation Army, Wuxi 214044, China ; 2 Department of Medical Oncology, 3 Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| |
Collapse
|
26
|
Olbryt M, Habryka A, Student S, Jarząb M, Tyszkiewicz T, Lisowska KM. Global gene expression profiling in three tumor cell lines subjected to experimental cycling and chronic hypoxia. PLoS One 2014; 9:e105104. [PMID: 25122487 PMCID: PMC4133353 DOI: 10.1371/journal.pone.0105104] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 07/18/2014] [Indexed: 11/19/2022] Open
Abstract
Hypoxia is one of the most important features of the tumor microenvironment, exerting an adverse effect on tumor aggressiveness and patient prognosis. Two types of hypoxia may occur within the tumor mass, chronic (prolonged) and cycling (transient, intermittent) hypoxia. Cycling hypoxia has been shown to induce aggressive tumor cell phenotype and radioresistance more significantly than chronic hypoxia, though little is known about the molecular mechanisms underlying this phenomenon. The aim of this study was to delineate the molecular response to both types of hypoxia induced experimentally in tumor cells, with a focus on cycling hypoxia. We analyzed in vitro gene expression profile in three human cancer cell lines (melanoma, ovarian cancer, and prostate cancer) exposed to experimental chronic or transient hypoxia conditions. As expected, the cell-type specific variability in response to hypoxia was significant. However, the expression of 240 probe sets was altered in all 3 cell lines. We found that gene expression profiles induced by both types of hypoxia were qualitatively similar and strongly depend on the cell type. Cycling hypoxia altered the expression of fewer genes than chronic hypoxia (6,132 vs. 8,635 probe sets, FDR adjusted p<0.05), and with lower fold changes. However, the expression of some of these genes was significantly more affected by cycling hypoxia than by prolonged hypoxia, such as IL8, PLAU, and epidermal growth factor (EGF) pathway-related genes (AREG, HBEGF, and EPHA2). These transcripts were, in most cases, validated by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Our results indicate that experimental cycling hypoxia exerts similar, although less intense effects, on the examined cancer cell lines than its chronic counterpart. Nonetheless, we identified genes and molecular pathways that seem to be preferentially regulated by cyclic hypoxia.
Collapse
Affiliation(s)
- Magdalena Olbryt
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
- * E-mail:
| | - Anna Habryka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Sebastian Student
- Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
| | - Michał Jarząb
- III Department of Radiation Therapy and Chemotherapy, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Tomasz Tyszkiewicz
- Nuclear Medicine and Endocrine Oncology Department, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Katarzyna Marta Lisowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Gliwice, Poland
| |
Collapse
|
27
|
Stanniocalcin-1 promotes metastasis in a human breast cancer cell line through activation of PI3K. Clin Exp Metastasis 2014; 31:787-94. [PMID: 25056605 DOI: 10.1007/s10585-014-9668-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 07/07/2014] [Indexed: 01/22/2023]
Abstract
Stanniocalcin-l (STC-1) is a secreted glycoprotein hormone that regulates calcium and phosphate homeostasis. STC-1 expression is upregulated in several cancers including breast cancer, and has been shown to be prognostic. Although these clinical observations implicate STC-1 as a potential tumor marker, it is still unclear whether STC-1 confers a malignant phenotype. In this study, this question was addressed by overexpressing STC-1 in the human breast cancer cell line MDA-MB-231 and examining the resultant phenotype in vitro and in vivo. Overexpression of STC-1 enhanced invasiveness of MDA-MB-231 cells in vitro and promoted their lung metastasis in vivo, while having no effect on proliferation, adhesion, or proteinase activity. The addition of soluble STC-1 to MDA-MB-231 cultures resulted in the activation of the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, suggesting a mechanistic basis for the observed increases in cell motility and metastasis. Taken together, it was indicated that secreted STC-1 promotes metastatic potential of breast cancer cells via activation of PI3K/AKT.
Collapse
|
28
|
Expression of stanniocalcin-1 and stanniocalcin-2 in laryngeal squamous cell carcinoma and correlations with clinical and pathological parameters. PLoS One 2014; 9:e95466. [PMID: 24743310 PMCID: PMC3990672 DOI: 10.1371/journal.pone.0095466] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/27/2014] [Indexed: 12/14/2022] Open
Abstract
Background Stanniocalcin-1 (STC1) and stanniocalcin-2 (STC2) are secreted glycoprotein hormones involved in various types of human malignancies. The roles of STC1 and STC2 in laryngeal squamous cell carcinoma (LSCC) remain unknown. We investigated correlations between STC1 and STC2 expression and clinicopathological or prognostic factors in LSCC. Methods Pre-surgical peripheral blood samples were collected between 2012 and 2013 from 62 patients with LSCC. Quantitative RT-PCR analysis was performed to examine mRNA levels of STC1 and STC2. Immunohistochemistry was performed to retrospectively analyze 90 paraffin-embedded LSCC tissue samples, which were obtained from patients who received surgery between 2006 and 2009. These patients did not have histories of treatment or malignancies. Univariate analysis of patient survival was performed by the Kaplan–Meier method. Multivariate analyses were performed with the Cox proportional hazards model. Results The relative mRNA levels of STC1 and STC2 in peripheral blood were significantly greater in LSCC patients than those of healthy volunteers (both P<0.05). STC2 protein expression in tumor tissues was associated with invasion into the thyroid cartilage, T-Stage, lymphatic metastasis, clinical stage, and pathological differentiation (all P<0.05). In addition, STC2 protein expression was an independent prognostic factor for overall survival in patients with LSCC (P = 0.025). In contrast, STC1 expression only correlated with clinical stage (P = 0.026) and was not an independent or significant prognostic factor. Conclusions Circulating STC1 and STC2 mRNA are potentially useful blood markers for LSCC. Our results strongly suggest that the STC2 protein, but not STC1, may be a valuable biomarker for LSCC malignancies and a prognostic marker for poor outcome following surgery. Future studies should examine STC2 as a novel molecular target for the treatment of LSCC.
Collapse
|
29
|
Molecular alterations and clinical relevance in esophageal squamous cell carcinoma. Front Med 2013; 7:401-10. [PMID: 24002746 DOI: 10.1007/s11684-013-0286-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 07/10/2013] [Indexed: 02/08/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common types of gastrointestinal cancers, and the fourth leading cause of cancer-related deaths in China. Early detection and intervention in time may dramatically increase the survival of the patients by initiating treatment regimens during earlier stages of ESCC or even during precancerous stages. Molecular classification will be useful for subtyping esophageal tumors or precancerous lesions to improve current therapeutics or early intervention of the disease. In this review, we summarize the findings in investigating the molecular alterations and clinical relevance of ESCC.
Collapse
|
30
|
Lee CW, Hwang I, Park CS, Lee H, Park DW, Kang SJ, Lee SW, Kim YH, Park SW, Park SJ. Expression of stanniocalcin-1 in culprit coronary plaques of patients with acute myocardial infarction or stable angina. J Clin Pathol 2013; 66:787-91. [DOI: 10.1136/jclinpath-2013-201563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
31
|
Song H, Xu B, Yi J. Clinical significance of stanniocalcin-1 detected in peripheral blood and bone marrow of esophageal squamous cell carcinoma patients. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:35. [PMID: 22537917 PMCID: PMC3546458 DOI: 10.1186/1756-9966-31-35] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 04/26/2012] [Indexed: 11/10/2022]
Abstract
Background Stanniocalcin-1 (STC-1) is a potential marker of disseminated tumor cells (DTCs). The aim of this study was to examine STC-1 expression in peripheral blood (PB) and bone marrow (BM) of esophageal squamous cell carcinoma (ESCC) patients, and to evaluate its clinical significance. Methods A total of 85 ESCC patients treated with radical resection were enrolled in this study. Immunohistochemistry was used to detect STC-1 protein expression in ESCC tissues. Nested RT-PCR was used to detect STC-1 mRNA expression in PB and BM. Results There were 71 cases (83.5%) showed a higher level of STC-1 protein expression in tumor tissues than in adjacent normal tissues (P < 0.001). Furthermore, the frequencies of STC-1 mRNA expression detected in PB and BM were 37.6% (32/85) and 21.2% (18/85), respectively, and together increased sensitivity to 48.2% (41/85), which was much higher than that in patients with benign esophageal disease (5.0%, 2/40, P < 0.001). In addition, STC-1 mRNA expression either in PB or BM was correlated with lymph metastasis, advanced stage and adverse 2-year progression free survival (PFS). In a multivariate analysis using the Cox proportional hazard model, STC-1 expression in PB and/or BM was an independent unfavorable prognostic factor for ESCC, apart from lymph metastasis and clinical stage. Conclusions STC-1 mRNA expression is a reliable marker for detection of DTCs in PB and BM of ESCC patients, and STC-1-positive DTCs may be a promising tool for diagnosis and prognosis assessment in ESCC.
Collapse
Affiliation(s)
- Haizhu Song
- Department of Medical Oncology, Jinling Hospital, 305 East Zhongshan Road, Nanjing, 210002, PR China
| | | | | |
Collapse
|