1
|
Khatun M, Modhukur V, Piltonen TT, Tapanainen JS, Salumets A. Stanniocalcin Protein Expression in Female Reproductive Organs: Literature Review and Public Cancer Database Analysis. Endocrinology 2024; 165:bqae110. [PMID: 39186548 PMCID: PMC11398916 DOI: 10.1210/endocr/bqae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/24/2024] [Indexed: 08/28/2024]
Abstract
Stanniocalcin (STC) 1 and 2 serve as antihyperglycemic polypeptide hormones with critical roles in regulating calcium and phosphate homeostasis. They additionally function as paracrine and/or autocrine factors involved in numerous physiological processes, including female reproduction. STC1 and STC2 contribute to the pathophysiology of several diseases, including female infertility- and pregnancy-associated conditions, and even tumorigenesis of reproductive organs. This comprehensive review highlights the dynamic expression patterns and potential dysregulation of STC1 and STC2, restricted to female fertility, and infertility- and pregnancy-associated diseases and conditions, such as endometriosis, polycystic ovary syndrome (PCOS), abnormal uterine bleeding, uterine polyps, and pregnancy complications, like impaired decidualization, preeclampsia, and preterm labor. Furthermore, the review elucidates the role of dysregulated STC in the progression of cancers of the reproductive system, including endometrial, cervical, and ovarian cancers. Additionally, the review evaluates the expression patterns and prognostic significance of STC in gynecological cancers by utilizing existing public datasets from The Cancer Genome Atlas to help decipher the multifaceted roles of these pleiotropic hormones in disease progression. Understanding the intricate mechanisms by which STC proteins influence all these reviewed conditions could lead to the development of targeted diagnostic and therapeutic strategies in the context of female reproductive health and oncology.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Competence Centre on Health Technologies, 50411 Tartu, Estonia
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Obstetrics and Gynaecology, HFR—Cantonal Hospital of Fribourg and University of Fribourg, 79085 Fribourg, Switzerland
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Competence Centre on Health Technologies, 50411 Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 14152 Huddinge, Stockholm, Sweden
| |
Collapse
|
2
|
Aydin AA, Yildirim S. Stanniocalcin-2 expression in glioblastoma - A novel prognostic biomarker: An observational study. Medicine (Baltimore) 2024; 103:e38913. [PMID: 38996177 PMCID: PMC11245224 DOI: 10.1097/md.0000000000038913] [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: 05/23/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The objective of this study was to assess the prognostic relevance of Stanniocalcin-2 (STC2) expression, as determined via immunohistochemistry in tumor tissue, in a cohort of 83 patients diagnosed with glioblastoma who underwent maximal safe surgical resection followed by radiotherapy concurrent with adjuvant temozolomide. STC2 expression levels were categorized using a 3-tiered semiquantitative system: negative expression (level 0-), low expression (level 1+), and high expression (levels 2 + and 3+). Patients were categorized into 2 distinct groups according to their STC2 expression levels: negative STC2 (-/+) and positive STC2 (++/+++). The primary outcome measure was the relationship between STC2 expression and progression-free survival (PFS), with overall survival (OS) serving as the secondary endpoint. Kaplan-Meier survival analysis confirmed that patients exhibiting high STC2 expression had significantly shorter OS (8 vs 20 months, P < .001) and PFS (6 vs 18 months, P < .001) than those with low or negative STC2 expression. Multivariate analysis revealed that STC2 expression was an independent prognostic factor for both OS (hazard ratio: 0.4; 95% confidence interval: 0.2-0.8; P < .05) and PFS (hazard ratio: 0.3; 95% confidence interval: 0.2-0.4; P < .05) in patients with glioblastoma. Furthermore, elevated STC2 expression in GBM was correlated with several established aggressive clinicopathological characteristics, including advanced age (≥65 years), low ECOG PS (≥2), and isocitrate dehydrogenase mutation negativity. These findings underscore that heightened STC2 expression within the tumor tissue of GBM patients functions as an adverse prognostic marker, correlating with an elevated risk of progression and reduced OS. Therapeutic interventions targeting the AKT-mTOR, ERK1-2, and mitogen-activated protein kinase pathways as well as immune checkpoint inhibitors and vascular endothelial growth factor blockade, as well as potential forthcoming antibody-drug conjugates targeting the STC2 molecule, have the potential to broaden the scope of combined treatment strategies.
Collapse
Affiliation(s)
- Asim Armagan Aydin
- Department of Clinical Oncology, Health Science University Antalya Training and Research Hospital, Antalya, Turkey
| | - Senay Yildirim
- Department of Pathology, Health Science University Antalya Training and Research Hospital, Antalya, Turkey
| |
Collapse
|
3
|
Zhong R, Zhan J, Zhang S. Integrative Analysis Reveals STC2 as a Prognostic Biomarker of Laryngeal Squamous Cell Carcinoma. Appl Biochem Biotechnol 2024; 196:3891-3913. [PMID: 37792175 DOI: 10.1007/s12010-023-04727-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/05/2023]
Abstract
Stanniocalcin 2 (STC2) is involved in many tumour types, but it remains unclear what its biological function is in laryngeal squamous cell carcinoma (LSCC). Therefore, we investigated STC2's expression, potential function, and prognostic significance of in LSCC. The expression and prognosis of STC2 in LSCC were described using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. In the TCGA database, the relationship between STC2 and immune infiltration, expression of immune cell chemokine and receptor genes, immune cell molecular marker genes, and epithelial‒mesenchymal transition (EMT) marker genes were analysed. The biological processes involved in STC2 and its expression-related genes were analysed comprehensively using bioinformatics. The single-gene ceRNA network of STC2 was constructed in the TCGA database. Finally, LSCC patients' tumour tissue STC2 expression was verified. STC2 silencing with the RNAi technique was used for the determination of cellular functions in a laryngeal cancer cell line. STC2 expression was higher in most tumours, including LSCC, than in normal tissues and was associated with poor prognosis. The relative proportions of naïve B, plasma, follicular helper T, and macrophage M0 cells in LSCC and normal samples differed significantly. STC2 expression correlated significantly positively with that of TGFB1 (biomarker of Tregs) and significantly negatively with that of D79A and CD19 (biomarkers of B cells). Furthermore, STC2 affected chemokine and receptor gene expression in immune cells. STC2 expression correlated with EMT marker gene expression in LSCC. STC2 was enriched in the PI3K/AKT signalling pathway, extracellular matrix (ECM) organisation, ECM-receptor interaction, and other tumour-related signalling pathways. STC2 was highly expressed in our clinical samples. N-cadherin and vimentin expression were decreased in the TU686 cell line after successful silencing of STC2, indicating that high STC2 expression may prompt LSCC cells to adopt a mesenchymal cell phenotype. STC2 silencing substantially reduced proliferation and migration in the TU686 cell line. STC2 may be a promising predictive biomarker for tumours, providing new approaches for LSCC diagnosis and treatment monitoring.
Collapse
Affiliation(s)
- Rong Zhong
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiandong Zhan
- Department of Otorhinolaryngology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Siyi Zhang
- School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Otorhinolaryngology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
4
|
Bu Q, Deng Y, Wang Q, Deng R, Hu S, Pei Z, Zhang Y. STC2 is a potential biomarker of hepatocellular carcinoma with its expression being upregulated in Nrf1α-deficient cells, but downregulated in Nrf2-deficient cells. Int J Biol Macromol 2023; 253:127575. [PMID: 37866563 DOI: 10.1016/j.ijbiomac.2023.127575] [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: 07/10/2023] [Revised: 10/03/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Nrf1 (encoded by Nfe2l1) and Nrf2 (encoded by Nfe2l2), as two key members of the CNC-bZIP transcription factor, exhibit significant functional differences in their pathophysiology. Our previous findings demonstrated that loss of Nrf1α (i.e., a full-length isoform of Nrf1) promotes HepG2-derived tumor growth in xenograft mice, but malgrowth of the xenograft tumor is significantly suppressed by knockout of Nrf2. To gain insights into the mechanism underlying such marked distinctions in their pathologic phenotypes, we mined transcriptome data from liver cancer in the TCGA database to establish a prognostic model and calculate predicted risk scores for each cell line. The results revealed that knockout of Nrf1α markedly increased the risk score in HepG2 cells, whereas the risk score was reduced by knockout of Nrf2. Notably, stanniocalcin 2 (STC2), a biomarker associated with liver cancer, that is upexpressed in hepatocellular carcinoma (HCC) tissues with a reduction in the overall survival ratio of those patients. We observed increased expression levels of STC2 in Nrf1α-/- cells but decreased expression in Nrf2-/- cells. These findings suggested that STC2 may play a role in mediating the distinction between Nrf1α-/- and Nrf2-/-. Such potential function of STC2 was further corroborated through a series of experiments combined with transcriptomic sequencing. The results revealed that STC2 functions as a dominant tumor-promoter, because the STC2-leading increases in clonogenicity of hepatoma cells and malgrowth of relevant xenograft tumor were almost completely abolished in STC2-/- cells. Together, these demonstrate that STC2 could be paved as a potential therapeutic target, albeit as a diagnostic marker, for HCC.
Collapse
Affiliation(s)
- Qiqi Bu
- Bioengineering College, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Yangxu Deng
- Bioengineering College, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Qing Wang
- Bioengineering College, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Rongzhen Deng
- Bioengineering College, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Shaofan Hu
- Bioengineering College, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Zhigang Pei
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China.
| |
Collapse
|
5
|
Chu H, Xie W, Guo C, Shi H, Gu J, Qin Z, Xie Y. Inhibiting stanniocalcin 2 reduces sunitinib resistance of Caki-1 renal cancer cells under hypoxia condition. Ann Med Surg (Lond) 2023; 85:5963-5971. [PMID: 38098599 PMCID: PMC10718379 DOI: 10.1097/ms9.0000000000001450] [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: 08/07/2023] [Accepted: 10/20/2023] [Indexed: 12/17/2023] Open
Abstract
Background Our previous study has suggested that blocking stanniocalcin 2 (STC2) could reduce sunitinib resistance in clear cell renal cell carcinoma (ccRCC) under normoxia. The hypoxia is a particularly important environment for RCC occurrence and development, as well as sunitinib resistance. The authors proposed that STC2 also plays important roles in RCC sunitinib resistance under hypoxia conditions. Methods The ccRCC Caki-1 cells were treated within the hypoxia conditions. Real-time quantitative PCR and Western blotting were applied to detect the STC2 expression in ccRCC Caki-1 cells. STC2-neutralizing antibodies, STC2 siRNA, and the recombinant human STC2 (rhSTC2) were used to identify targeting regulation on STC2 in modulating sunitinib resistance, proliferation, epithelial-mesenchymal transition (EMT), migration, and invasion. In addition, autophagy flux and the lysosomal acidic environment were investigated by Western blotting and fluorescence staining, and the accumulation of sunitinib in cells was observed with the addition of STC2-neutralizing antibodies and autophagy modulators. Results Under hypoxia conditions, sunitinib disrupted the lysosomal acidic environment and accumulated in Caki-1 cells. Hypoxia-induced the STC2 mRNA and protein levels in Caki-1 cells. STC2-neutralizing antibodies and STC2 siRNA effectively aggravated sunitinib-reduced cell viability and proliferation, which were reversed by rhSTC2. In addition, sunitinib promoted EMT, migration, and invasion, which were reduced by STC2-neutralizing antibodies. Conclusion Inhibiting STC2 could reduce the sunitinib resistance of ccRCC cells under hypoxia conditions.
Collapse
Affiliation(s)
- Hezhen Chu
- Department of Urology, Yixing Traditional Chinese Medicine Hospital
| | - Wenchao Xie
- Department of Urology, Affiliated Hospital of Jiangsu University-Yixing People’s Hospital, Yixing
| | - Chuanzhi Guo
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Zhenqian Qin
- Department of Urology, Affiliated Hospital of Jiangsu University-Yixing People’s Hospital, Yixing
| | - Yimin Xie
- Department of Urology, Affiliated Hospital of Jiangsu University-Yixing People’s Hospital, Yixing
| |
Collapse
|
6
|
He C, Zhang G, Lu Y, Zhou J, Ren Z. DDX17 modulates the expression and alternative splicing of genes involved in apoptosis and proliferation in lung adenocarcinoma cells. PeerJ 2022; 10:e13895. [PMID: 36164607 PMCID: PMC9508879 DOI: 10.7717/peerj.13895] [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: 04/11/2022] [Accepted: 07/22/2022] [Indexed: 01/19/2023] Open
Abstract
Background The DEAD-box RNA-binding protein (RBP) DDX17 has been found to be involved in the tumorigenesis of many types of cancers. However, the role of DDX17 in lung adenocarcinoma (LUAD) remains unclear. Methods We silenced DDX17 expression in A549 LUAD cells by small interfering RNA (siRNA). Cell proliferation and apoptosis assays were performed to explore the functions of DDX17. Knockdown of DDX17 by siRNA significantly inhibited proliferation and induced apoptosis in A549 cells. We used high-throughput RNA sequencing (RNA-seq) to identify differentially expressed genes (DEGs) and alternative splicing (AS) events in DDX17 knockdown LUAD cells. Results DDX17 knockdown increased the expression levels of proapoptotic genes and decreased those of proproliferative genes. Moreover, the DDX17-regulated AS events in A549 cells revealed by computational analysis using ABLas software were strongly validated by quantitative reverse transcription-polymerase chain reaction (RT-qPCR) and were also validated by analysis of The Cancer Genome Atlas (TCGA)-LUAD dataset. These findings suggest that DDX17 may function as an oncogene by regulating both the expression and AS of proliferation- and apoptosis-associated genes in LUAD cells. Our findings may offer new insights into understanding the molecular mechanisms of LUAD and provide a new therapeutic direction for LUAD.
Collapse
Affiliation(s)
- Cheng He
- Department of Thoracic Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China,Department of Thoracic Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Gan Zhang
- Department of Thoracic Surgery, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Yanhong Lu
- Department of Thoracic Surgery, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Jingyue Zhou
- Department of Thoracic Surgery, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Zixue Ren
- Department of Thoracic Surgery, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| |
Collapse
|
7
|
Yu L, Liu X, Wang X, Yan H, Pu Q, Xie Y, Du J, Yang Z. Glycometabolism-related gene signature of hepatocellular carcinoma predicts prognosis and guides immunotherapy. Front Cell Dev Biol 2022; 10:940551. [PMID: 35938165 PMCID: PMC9354664 DOI: 10.3389/fcell.2022.940551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a severe cancer endangering human health. We constructed a novel glycometabolism-related risk score to predict prognosis and immunotherapy strategies in HCC patients. The HCC data sets were obtained from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database, and the glycometabolism-related gene sets were obtained from the Molecular Signature Database. The least absolute contraction and selection operator (LASSO) regression model was used to construct a risk score based on glycometabolism-related genes. A simple visual nomogram model with clinical indicators was constructed and its effectiveness in calibration, accuracy, and clinical value was evaluated. We also explored the correlation between glycometabolism-related risk scores and molecular pathways, immune cells, and functions. Patients in the low-risk group responded better to anti-CTLA-4 immune checkpoint treatment and benefited from immune checkpoint inhibitor (ICI) therapy. The study found that glycometabolism-related risk score can effectively distinguish the prognosis, molecular and immune-related characteristics of HCC patients, and may provide a new strategy for individualized treatment.
Collapse
Affiliation(s)
- Lihua Yu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Liu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xinhui Wang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Huiwen Yan
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Qing Pu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuqing Xie
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Juan Du
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhiyun Yang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Birzniece V, Lam T, McLean M, Reddy N, Shahidipour H, Hayden A, Gurney H, Stone G, Hjortebjerg R, Frystyk J. Insulin-like growth factor role in determining the anti-cancer effect of metformin: RCT in prostate cancer patients. Endocr Connect 2022; 11:EC-21-0375. [PMID: 35324467 PMCID: PMC9066575 DOI: 10.1530/ec-21-0375] [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: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Androgen deprivation therapy (ADT), a principal therapy in patients with prostate cancer, is associated with the development of obesity, insulin resistance, and hyperinsulinemia. Recent evidence indicates that metformin may slow cancer progression and improves survival in prostate cancer patients, but the mechanism is not well understood. Circulating insulin-like growth factors (IGFs) are bound to high-affinity binding proteins, which not only modulate the bioavailability and signalling of IGFs but also have independent actions on cell growth and survival. The aim of this study was to investigate whether metformin modulates IGFs, IGF-binding proteins (IGFBPs), and the pregnancy-associated plasma protein A (PAPP-A) - stanniocalcin 2 (STC2) axis. DESIGN AND METHODS In a blinded, randomised, cross-over design, 15 patients with prostate cancer on stable ADT received metformin and placebo treatment for 6 weeks each. Glucose metabolism along with circulating IGFs and IGFBPs was assessed. RESULTS Metformin significantly reduced the homeostasis model assessment as an index of insulin resistance (HOMA IR) and hepatic insulin resistance. Metformin also reduced circulating IGF-2 (P < 0.05) and IGFBP-3 (P < 0.01) but increased IGF bioactivity (P < 0.05). At baseline, IGF-2 correlated significantly with the hepatic insulin resistance (r2= 0.28, P < 0.05). PAPP-A remained unchanged but STC2 declined significantly (P < 0.05) following metformin administration. During metformin treatment, change in HOMA IR correlated with the change in STC2 (r2= 0.35, P < 0.05). CONCLUSION Metformin administration alters many components of the circulating IGF system, either directly or indirectly via improved insulin sensitivity. Reduction in IGF-2 and STC2 may provide a novel mechanism for a potential metformin-induced antineoplastic effect.
Collapse
Affiliation(s)
- Vita Birzniece
- School of Medicine, Western Sydney University, New South Wales, Australia
- Department of Diabetes and Endocrinology, Blacktown Hospital, New South Wales, Australia
- Garvan Institute of Medical Research, New South Wales, Australia
- School of Medical Sciences, University of New South Wales, New South Wales, Australia
- Correspondence should be addressed to V Birzniece:
| | - Teresa Lam
- School of Medicine, Western Sydney University, New South Wales, Australia
- Department of Diabetes and Endocrinology, Blacktown Hospital, New South Wales, Australia
- Department of Diabetes and Endocrinology, Westmead Hospital, New South Wales, Australia
| | - Mark McLean
- School of Medicine, Western Sydney University, New South Wales, Australia
- Department of Diabetes and Endocrinology, Blacktown Hospital, New South Wales, Australia
| | - Navneeta Reddy
- Department of Diabetes and Endocrinology, Blacktown Hospital, New South Wales, Australia
| | - Haleh Shahidipour
- School of Medicine, Western Sydney University, New South Wales, Australia
- Department of Diabetes and Endocrinology, Blacktown Hospital, New South Wales, Australia
| | - Amy Hayden
- School of Medicine, Western Sydney University, New South Wales, Australia
- Faculty of Medicine, Health and Human Sciences, Macquarie University, New South Wales, Australia
- Crown Princess Mary Cancer Centre, Westmead Hospital, New South Wales, Australia
| | - Howard Gurney
- Crown Princess Mary Cancer Centre, Westmead Hospital, New South Wales, Australia
| | - Glenn Stone
- School of Computing, Engineering and Mathematics, Western Sydney University, New South Wales, Australia
| | - Rikke Hjortebjerg
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital & Department of Clinical Research, Faculty of Health, University of Southern Denmark, Odense, Denmark
| | - Jan Frystyk
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
9
|
Wu Z, Cheng H, Liu J, Zhang S, Zhang M, Liu F, Li Y, Huang Q, Jiang Y, Chen S, Lv L, Li D, Zeng JZ. The Oncogenic and Diagnostic Potential of Stanniocalcin 2 in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2022; 9:141-155. [PMID: 35300206 PMCID: PMC8922464 DOI: 10.2147/jhc.s351882] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/26/2022] [Indexed: 02/05/2023] Open
Abstract
Purpose Early detection and prognostic prediction of hepatocellular carcinoma (HCC) remain a great challenge. In this study, we explored the role and diagnostic significance of stanniocalcin 2 (STC2), recently identified as a secretory protein, in HCC. Methods STC2 mRNA and protein in HCC tissues were examined by qRT-PCR and immunohistochemistry. The regulatory role of HCC growth by STC2 was evaluated in vitro and in vivo. Serum STC2 levels were determined in HCC patients and compared to those with liver cirrhosis (LC) and normal controls (NC). The difference and significance of STC2 levels between groups were analyzed by Mann–Whitney U-test. The diagnostic value of serum STC2 in detecting early HCC was assayed with receiver operating characteristics (ROC). The association of STC2 with overall survival (OS) was determined with Kaplan–Meier method. Results STC2 was elevated in about 77.1% HCC patients and correlated with advanced tumor progression. Overexpression or knockdown of STC2 stimulated or suppressed HCC colony formation and xenograft tumor growth. AKT activation played a critical role in tumor-promoting effect of STC2. The median level of serum STC2 in HCC patients (n = 98, 2086.6 ng/L) was 2.6-fold and 4.2-fold that in LC patients (n = 42, 801.9 ng/L) and NC (n = 26, 496.9 ng/L), respectively. A cut-off value 1493 ng/L for STC2 could distinguish early HCC from LC with a sensitivity of 76.9% and a specificity of 76.2%, both of which were superior to AFP at 20 μg/L (sensitivity 69.2%, specificity 52.4%). STC2 was positive in 77.8% (14/18) AFP-negative patients. High STC2 level was correlated with poor overall and disease specific survival. Conclusion STC2 is upregulated in both tumor and serum of HCC patients, and its overexpression promotes HCC via AKT pathway. STC2 possesses a diagnostic significance and may serve as an auxiliary biomarker of AFP for detecting early HCC.
Collapse
Affiliation(s)
- Zhixian Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Hongwei Cheng
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
| | - Jie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
| | - Shuaishuai Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
| | - Minda Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
| | - Fangzhou Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
| | - Yinghui Li
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Qian Huang
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Yi Jiang
- Department of Hepatobiliary Surgery, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Shaohua Chen
- Department of Hepatobiliary Surgery, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Lizhi Lv
- Department of Hepatobiliary Surgery, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Dongliang Li
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, People’s Republic of China
| | - Jin-Zhang Zeng
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, People’s Republic of China
- Correspondence: Jin-Zhang Zeng; Dongliang Li, Email ;
| |
Collapse
|
10
|
Li N, Su M, Zhu L, Wang L, Peng Y, Dong B, Ma L, Liu Y. A Prognostic Signature of Glycolysis-Related Long Noncoding RNAs for Molecular Subtypes in the Tumor Immune Microenvironment of Lung Adenocarcinoma. Int J Gen Med 2021; 14:8955-8974. [PMID: 34866936 PMCID: PMC8637177 DOI: 10.2147/ijgm.s340615] [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: 09/23/2021] [Accepted: 11/10/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Long noncoding RNAs (lncRNAs) and glycolysis regulate multiple types of cancer. However, the prognostic roles and biological functions of glycolysis-related lncRNAs in lung adenocarcinoma (LUAD) remain unclear. In this study, we investigated the role of glycolysis-related lncRNAs in LUAD. Patients and Methods We retrieved glycolysis-related genes from the Molecular Signatures Database and screened for prognostic glycolysis-related lncRNAs from The Cancer Genome Atlas. Results We identified three LUAD subtypes (clusters 1–3) by univariate Cox regression analysis and consensus clustering. Patients in cluster 1 had the best overall survival rates. Immune, stromal, and cytolytic-activity scores were the highest in cluster 1. The expression of immune checkpoint molecules (programmed cell death protein 1 and cytotoxic T-lymphocyte-associated protein 4) and other immune-related indicators was the highest in cluster 1, whereas that of epithelial cell biomarkers (Cadherin 1, Cadherin 2, and MET) was the lowest. Therefore, patients in cluster 1 may benefit from immunotherapy. Lasso–Cox regression and multivariate Cox regression analyses were used to select nine lncRNAs to build a robust prognostic model of LUAD. The area under the curve classifier values and a nomogram performed well in predicting survival times for patients with LUAD. The expression levels of nine lncRNAs were validated by quantitative reverse transcriptase-polymerase chain reaction analysis, and most of these lncRNAs were significantly related to immune-related mRNAs. Gene set enrichment analysis revealed that the high-risk group was enriched for cell cycle-related pathways and the low-risk group was enriched for pathways associated with immunity or immune-related diseases. Conclusion The LUAD subtypes and prognostic model developed here may help in clinical risk stratification, prognosis management, and treatment decisions for patients with LUAD.
Collapse
Affiliation(s)
- Na Li
- Department of Central Laboratory, Shenyang Tenth People's Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, People's Republic of China
| | - Mu Su
- Berry Oncology Corporation, Beijing, People's Republic of China
| | - Louyin Zhu
- Berry Oncology Corporation, Beijing, People's Republic of China
| | - Li Wang
- Berry Oncology Corporation, Beijing, People's Republic of China
| | - Yonggang Peng
- Berry Oncology Corporation, Beijing, People's Republic of China
| | - Bo Dong
- Department of Central Laboratory, Shenyang Tenth People's Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, People's Republic of China
| | - Liya Ma
- Department of Central Laboratory, Shenyang Tenth People's Hospital, Shenyang Chest Hospital, Shenyang, Liaoning, People's Republic of China
| | - Yongyu Liu
- Department of Thoracic Surgery, Shenyang Tenth People's Hospital, Shenyang Chest Hospital, Shenyang, 110044, Liaoning, People's Republic of China
| |
Collapse
|
11
|
Li S, Huang Q, Li D, Lv L, Li Y, Wu Z. The significance of Stanniocalcin 2 in malignancies and mechanisms. Bioengineered 2021; 12:7276-7285. [PMID: 34612765 PMCID: PMC8806499 DOI: 10.1080/21655979.2021.1977551] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Human stanniocalcin 2 (STC2) is an ortholog of fish stanniocalcins (STCs) and is widely expressed in various organs and tissues. The gene is localized on chromosome 5q33 or 5q35. STC2 has been implicated in glucose homeostasis and phosphorus metabolism. It is also reported to be implicated in various malignancies. STC2 was found to be implicated in breast cancer and gynecologic cancers, suggesting hormone-specific or -dependent activities in these malignancies. Moreover, it was reported to be involved in gastrointestinal tumors, including esophageal, gastric, colorectal, and liver cancers, and respiratory cancers, including laryngeal and lung cancers. It also influenced renal carcinoma and prostate cancer. Notably, as a secreted phosphoprotein, STC2 was detectable in serum and possessed promising predictive value in several malignancies. This review aims to improve the understanding of the role of STC2 in patient diagnosis and prognosis, and tumor development and progression, as well as the mechanisms involved.
Collapse
Affiliation(s)
- Shasha Li
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Qian Huang
- Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Dongliang Li
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Lizhi Lv
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| | - Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Zhixian Wu
- Department of Hepatobiliary Disease, Dongfang Hospital, Xiamen University, Fuzhou, China.,Department of Hepatobiliary Disease, Fuzong Clinical College, Fujian Medical University, Fuzhou, China
| |
Collapse
|
12
|
Zhang Y, Chen P, Zhou Q, Wang H, Hua Q, Wang J, Zhong H. A Novel Immune-Related Prognostic Signature in Head and Neck Squamous Cell Carcinoma. Front Genet 2021; 12:570336. [PMID: 34220923 PMCID: PMC8249947 DOI: 10.3389/fgene.2021.570336] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 04/20/2021] [Indexed: 02/05/2023] Open
Abstract
The immune response within the tumor microenvironment plays a key role in tumorigenesis and determines the clinical outcomes of head and neck squamous cell carcinoma (HNSCC). However, to date, very limited robust and reliable immunological biomarkers have been developed that are capable of estimating prognosis in HNSCC patients. In this study, we aimed to identify the effects of novel immune-related gene signatures (IRGs) that can predict HNSCC prognosis. Based on gene expression profiles and clinical data of HNSCC patient cohorts from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database, a total of 439 highly variable expressed immune-related genes (including 239 upregulated and 200 downregulated genes) were identified by using differential gene expression analysis. Pathway enrichment analysis indicated that these immune-related differentially expressed genes were enriched in inflammatory functions. After process screening in the training TCGA cohort, six immune-related genes (PLAU, STC2, TNFRSF4, PDGFA, DKK1, and CHGB) were significantly associated with overall survival (OS) based on the LASSO Cox regression model. Integrating these genes with clinicopathological features, a multivariable model was built and suggested better performance in determining patients’ OS in the testing cohort, and the independent validation cohort. In conclusion, a well-established model encompassing both immune-related gene signatures and clinicopathological factors would serve as a promising tool for the prognostic prediction of HNSCC.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiang Zhou
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongyan Wang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongliang Zhong
- Department of Neurosurgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Li T, Feng Z, Wang Y, Zhang H, Li Q, Schiferle E, Qin Y, Xiao S. Antioncogenic Effect of MicroRNA-206 on Neck Squamous Cell Carcinoma Through Inhibition of Proliferation and Promotion of Apoptosis and Autophagy. Hum Gene Ther 2020; 31:1260-1273. [PMID: 32900244 DOI: 10.1089/hum.2020.090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Recent studies have reported the crucial role of stanniocalcin-2 (STC2) in hepatocellular carcinoma; however, its role in head and neck squamous cell carcinoma (HNSCC) remains elusive. In this study, microRNA-206 (miR-206) was predicted to target STC2 gene. The study herein aimed to elucidate the effect of miR-206 on HNSCC by targeting STC2. STC2 was highly expressed in HNSCC tissues and cells. By targeting STC2, miR-206 decreased mRNA and protein expression of STC2. Importantly, our study showed that miR-206 blocked the Akt signaling pathway by inhibiting STC2. Intriguingly, our data from in vitro and in vivo experiments suggested that miR-206 overexpression led to decreased cell proliferation and increased cell apoptosis and autophagy, as well as suppressed tumor growth; whereas, STC2 silencing reversed the effects of miR-206 inhibitor on those biological behaviors. In this study, we investigated the antioncogenic effect of miR-206 on HNSCC by targeting STC2, and highlighted miR-206/STC2 aixs as potential therapeutic targets for HNSCC.
Collapse
Affiliation(s)
- Tiancheng Li
- Departments of Otorhinolaryngology-Head and Neck Surgery ,Peking University First Hospital, Beijing, P.R. China
| | - Zhien Feng
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, P.R. China
| | - Yingyi Wang
- Department of Oncology, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Hong Zhang
- Departments of Pathology, Peking University First Hospital, Beijing, P.R. China
| | - Qian Li
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Erik Schiferle
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yao Qin
- Departments of Otorhinolaryngology-Head and Neck Surgery ,Peking University First Hospital, Beijing, P.R. China
| | - Shuifang Xiao
- Departments of Otorhinolaryngology-Head and Neck Surgery ,Peking University First Hospital, Beijing, P.R. China
| |
Collapse
|
14
|
Xie F, Bai Y, Yang X, Long J, Mao J, Lin J, Wang D, Song Y, Xun Z, Huang H, Yang X, Zhang L, Mao Y, Sang X, Zhao H. Comprehensive analysis of tumour mutation burden and the immune microenvironment in hepatocellular carcinoma. Int Immunopharmacol 2020; 89:107135. [PMID: 33189609 DOI: 10.1016/j.intimp.2020.107135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/19/2020] [Accepted: 10/20/2020] [Indexed: 12/31/2022]
Abstract
Tumour mutation burden (TMB) and the immune microenvironment (IME) are reportedly associated with immunotherapy responses, but this relationship remains unclear in hepatocellular carcinoma (HCC). We classified HCC patients in the liver hepatocellular carcinoma cohort from The Cancer Genome Atlas into low- and high-TMB groups and evaluated differences in immune infiltrates. Additionally, differentially expressed genes in the low- and high-TMB groups were identified, and functional analyses were conducted. A risk score model was constructed based on three differentially expressed immune genes (DEIGs). The Tumor Immune Estimation Resource database was utilized to analyse how the IME was affected by the three hub DEIGs. Finally, a prognostic nomogram combining risk scores and stages was established and externally validated with the International Cancer Genome Consortium and GSE14520 cohorts. High-TMB (top 20%) patients exhibited a worse prognosis (P = 0.017). Follicular helper cells (P = 0.001) and activated natural killer cells (P = 0.003) were enriched in high-TMB patients, while resting dendritic cells (P = 0.002) were enriched in low-TMB samples. A risk score model was generated with three hub DEIGs (CCR7, STC2 and S100A9) to predict overall survival in HCC cohorts. Moreover, copy number variations mainly reduced infiltration levels. The nomogram performed better than the risk score model in the training and validation datasets. Higher TMB was associated with IME diversification and worse prognosis in HCC. Mutations in three hub TMB-associated DEIGs correlated with lower immune cell infiltration.
Collapse
Affiliation(s)
- Fucun Xie
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Yi Bai
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China; Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| | - Xu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Junyu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Jinzhu Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Jianzhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Dongxu Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Yang Song
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Ziyu Xun
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Hanchan Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Xiaobo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Lei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| |
Collapse
|
15
|
Zhang J, Bing Z, Yan P, Tian J, Shi X, Wang Y, Yang K. Identification of 17 mRNAs and a miRNA as an integrated prognostic signature for lung squamous cell carcinoma. J Gene Med 2020; 21:e3105. [PMID: 31215090 DOI: 10.1002/jgm.3105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Gene signatures for predicting the outcome of lung squamous cell carcinoma (LUSC) have been employed for many years. However, various signatures have been applied in clinical practice. Therefore, in the present study, we aimed to filter out an effective LUSC prognostic gene signature by simultaneously integrating mRNA and microRNA (miRNA). METHODS First, based on data from the Cancer Genome Atlas (TCGA) (https://www.cancer.gov/tcga), mRNAs and miRNAs that were related to overall survival of LUSC were obtained by the least absolute shrinkage and selection operator method. Subsequently, the predicting effect was tested by time-dependent receiver operating characteristic curve analysis and Kaplan-Meier survival analysis. Next, related clinical indices were added to evaluate the efficiency of the selected gene signatures. Finally, validation and comparison using three independent gene signatures were performed using data from the Gene Expression Omnibus database (https://www.ncbi.nlm.nih.gov/geo). RESULTS Our data showed that the prognostic index (PI) contained 17 mRNAs and one miRNA. According to the best normalized cut-off of PI (0.0247), the hazard ratio of the PI was 3.40 (95% confidence interval = 2.33-4.96). Moreover, when clinical factors were introduced, the PI was still the most significant index. In addition, only two Gene Ontology terms with p < 0.05 were reported. Furthermore, validation implied that, using our 18-gene signature, only hazard ratio = 1.36 (95% confidence interval = 1.01-1.83) was significant compared to the other three groups of gene biomarkers. CONCLUSIONS The 18-gene signature selected based on data from the TCGA database had an effective prognostic value for LUSC patients.
Collapse
Affiliation(s)
- Jingyun Zhang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Zhitong Bing
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Peijing Yan
- Institution of Clinical Research and Evidence Based Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Xiue Shi
- Gansu Rehabilitation Center Hospital, Lanzhou, China.,Gansu Evidence-Based Rehabilitation Medicine Center, Lanzhou, China
| | - Yongfeng Wang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Kehu Yang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,Institution of Clinical Research and Evidence Based Medicine, Gansu Provincial Hospital, Lanzhou, China.,Gansu Evidence-Based Rehabilitation Medicine Center, Lanzhou, China
| |
Collapse
|
16
|
Gu HY, Lin LL, Zhang C, Yang M, Zhong HC, Wei RX. The Potential of Five Immune-Related Prognostic Genes to Predict Survival and Response to Immune Checkpoint Inhibitors for Soft Tissue Sarcomas Based on Multi-Omic Study. Front Oncol 2020; 10:1317. [PMID: 32850416 PMCID: PMC7396489 DOI: 10.3389/fonc.2020.01317] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Low response rates to immunotherapy have been reported in soft tissue sarcoma (STS). There are few predictive biomarkers of response, and the tumor immune microenvironment associated with progression and prognosis remains unclear in STS. Gene expression data from the Cancer Genome Atlas were used to identify the immune-related prognostic genes (IRPGs) and construct the immune gene-related prognostic model (IGRPM). The tumor immune microenvironment was characterized to reveal differences between patients with different prognoses. Furthermore, somatic mutation data and DNA methylation data were analyzed to understand the underlying mechanism leading to different prognoses. The IGRPM was constructed using five IRPGs (IFIH1, CTSG, STC2, SECTM1, and BIRC5). Two groups (high- and low-risk patients) were identified based on the risk score. Low-risk patients with higher overall survival time had higher immune scores, more immune cell infiltration (e.g., CD8 T cell and activated natural killer cells), higher expression of immune-stimulating molecules, higher stimulating cytokines and corresponding receptors, higher innate immunity molecules, and stronger antigen-presenting capacity. However, inhibition of immunity was observed in low-risk patients owing to the higher expression of immune checkpoint molecules and inhibiting cytokines. High-risk patients had high tumor mutation burden, which did not significantly influence survival. Gene set enrichment analysis further revealed that pathways of cell cycle and cancers were activated in high-risk patients. DNA methylation analysis indicated that relative high methylation was associated with better overall survival. Finally, the age, mitotic counts, and risk scores were independent prognostic factors for STS. Five IRPGs performed well in risk stratification of patients and are candidate biomarkers for predicting response to immunotherapy. Differences observed through the multi-omic study of patients with different prognoses may reveal the underlying mechanism of the development and progression of STS, and thereby improve treatment.
Collapse
Affiliation(s)
- Hui-Yun Gu
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lu-Lu Lin
- Department of Pathology and Pathophysiology, School of Basic Medicine, Wuhan University, Wuhan, China
| | - Chao Zhang
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,Department of Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Min Yang
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hou-Cheng Zhong
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ren-Xiong Wei
- Department of Spine and Orthopedic Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Ferreira do Carmo A, Dourado MR, Ervolino de Oliveira C, Bastos DC, Domingueti CB, Ribeiro Paranaíba LM, Sawazaki-Calone Í, Borges GÁ, Silva Guerra EN, Casarin RC, Graner E, Salo TA, de Almeida Freitas R, Galvão HC, Coletta RD. Stanniocalcin 2 contributes to aggressiveness and is a prognostic marker for oral squamous cell carcinoma. Exp Cell Res 2020; 393:112092. [PMID: 32445747 DOI: 10.1016/j.yexcr.2020.112092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/09/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
Stanniocalcin 2 (STC2), a glycoprotein that regulates calcium and phosphate homeostasis during mineral metabolism, appears to display multiple roles in tumorigenesis and cancer progression. This study aimed to access the prognostic value of STC2 in oral squamous cell carcinoma (OSCC) and its implications in oral tumorigenesis. STC2 expression was examined in 2 independent cohorts of OSCC tissues by immunohistochemistry. A loss-of-function strategy using shRNA targeting STC2 was employed to investigate STC2 in vitro effects on proliferation, apoptosis, migration, invasion, epithelial-mesenchymal transition (EMT) and possible activation of signaling pathways. Moreover, STC2 effects were assessed in vivo in a xenograft mouse cancer model. High expression of STC2 was significantly associated with poor disease-specific survival (HR: 2.67, 95% CI: 1.37-5.21, p = 0.001) and high rate of recurrence with a hazard ratio of 2.80 (95% CI: 1.07-5.71, p = 0.03). In vitro downregulation of STC2 expression in OSCC cells attenuated proliferation, migration and invasiveness while increased apoptotic rates. In addition, the STC2 downregulation controlled EMT phenotype of OSCC cells, with regulation on E-cadherin, vimentin, Snail1, Twist and Zeb2. The reactivation of STC2 was observed in the STC2 knockdown cells in the in vivo xenograft model, and no influence on tumor growth was observed. Modulation of STC2 expression levels did not alter consistently the phosphorylation status of CREB, ERK, JNK, p38, p70 S6K, STAT3, STAT5A/B and AKT. Our findings suggest that STC2 overexpression is an independent marker of OSCC outcome and may contribute to tumor progression via regulation of proliferation, survival and invasiveness of OSCC cells.
Collapse
Affiliation(s)
- Andreia Ferreira do Carmo
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil; Department of Dentistry, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Mauricio Rocha Dourado
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil
| | - Carine Ervolino de Oliveira
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Débora Campanella Bastos
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil
| | - Catherine Bueno Domingueti
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Lívia Máris Ribeiro Paranaíba
- Department of Pathology and Parasitology, Institute of Biomedical Sciences, Federal University of Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Íris Sawazaki-Calone
- Oral Pathology and Oral Medicine, Dentistry School, Western Paraná; State University, Cascavel, Paraná, Brazil
| | - Gabriel Álvares Borges
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasilia, Brasilia, Brazil
| | - Eliete Neves Silva Guerra
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasilia, Brasilia, Brazil
| | - Renato C Casarin
- Department of Prosthodontics and Periodontics, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil
| | - Edgard Graner
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil
| | - Tuula A Salo
- Cancer and Translational Medicine Research Unit, Faculty of Medicine and Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland; Institute of Oral and Maxillofacial Disease, University of Helsinki, and HUSLAB, Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | | | - Hébel Cavalcanti Galvão
- Department of Dentistry, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba, SP, Brazil.
| |
Collapse
|
18
|
Joshi AD. New Insights Into Physiological and Pathophysiological Functions of Stanniocalcin 2. Front Endocrinol (Lausanne) 2020; 11:172. [PMID: 32296395 PMCID: PMC7136389 DOI: 10.3389/fendo.2020.00172] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stanniocalcin, a glycosylated peptide hormone, first discovered in a bony fish has originally been shown to play critical role in calcium and phosphate homeostasis. Two paralogs of stanniocalcin (STC1 and STC2) identified in mammals are widely expressed in variety of tissues. This review provides historical perspective on the discovery of fish and mammalian stanniocalcin, describes molecular regulation of STC2 gene, catalogs distribution as well as expression of STC2 in tissues, and provides key structural information known till date regarding mammalian STC2. Additionally, this mini review summarizes pivotal functions of STC2 in calcium and phosphate regulation, cytoprotection, cell development, and angiogenesis. Finally, STC2's role as a novel marker for human cancers has also been outlined. Reviewing these studies will provide an opportunity to understand STC2's structure, biological functions as well as key molecular pathways involving STC2, which will help us design innovative therapeutic interventions using this novel hormone.
Collapse
Affiliation(s)
- Aditya D. Joshi
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
19
|
Maciukiewicz M, Tiwari AK, Zai CC, Gorbovskaya I, Laughlin CP, Nurmi EL, Liebermann JA, Meltzer HY, Kennedy JL, Müller DJ. Genome-wide association study on antipsychotic-induced weight gain in Europeans and African-Americans. Schizophr Res 2019; 212:204-212. [PMID: 31447353 DOI: 10.1016/j.schres.2019.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 06/13/2019] [Accepted: 07/11/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Antipsychotic (AP) medications are the first line of treatment for schizophrenia. However, most conferr a risk of antipsychotic-induced weight gain (AIWG). The objective of this investigation was to conduct a genome-wide association study (GWAS) of AIWG, followed by comprehensive, post-GWAS approaches. METHODS We investigated n = 201 schizophrenia or schizoaffective disorder patients of European and African American ancestry who were treated primarily with clozapine or olanzapine. We conducted a genome-wide association analysis for AIWG, defined primarily as a percentage of weight change from baseline. RESULTS When examining Europeans (n = 147), we noticed an association between rs62097526 (β = 0.39, p = 3.59 × 10-6, CADD = 2.213) variant, located downstream of the CIDEA gene, which is considered a risk factor for AIWG. In the entire sample, we observed a significant association between rs1525085 (β = 0.411, p = 3.15 × 10-9) variant of the DGKB gene and AIWG. The association was nominally significant in Europeans (β = 0.271, p = 0.002) and African Americans (β = 0.579, p = 5.73 × 10-5) with the same risk allele. Our top genes (p < 5 × 10-5) were enriched in the GWAS catalog for the risk of obesity and interacted with the known risk factors for obesity (G6PD) and diabetes (IRS1). In addition, these genes are targeted by miRNAs related to schizophrenia (mir-34a) and obesity (mir-19b). However, our polygenic risk score analyses did not provide support for major genetic overlap between obesity and the risk of AIWG. CONCLUSIONS In summary, we propose that the CIDEA and DGKB genes are risk factors for AIWG in transethnic populations. Additionally, our evidence suggests that the G6PD and IRS1 gene-related pathways might be involved in AIWG.
Collapse
Affiliation(s)
- Malgorzata Maciukiewicz
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Arun K Tiwari
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Clement C Zai
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Ilona Gorbovskaya
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Christopher P Laughlin
- Division of Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience, Los Angeles, California, USA
| | - Erika L Nurmi
- Division of Child and Adolescent Psychiatry, UCLA Semel Institute for Neuroscience, Los Angeles, California, USA
| | - Jeffrey A Liebermann
- Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Herbert Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL, USA
| | - James L Kennedy
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J Müller
- Pharmacogenetic Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
STC2 Is a Potential Prognostic Biomarker for Pancreatic Cancer and Promotes Migration and Invasion by Inducing Epithelial-Mesenchymal Transition. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8042489. [PMID: 32258098 PMCID: PMC7099867 DOI: 10.1155/2019/8042489] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 01/20/2023]
Abstract
Aberrant expression of stanniocalcin 2 (STC2) is implicated in cancer development. STC2 acts as a tumor promoter to drive some cancers. However, its contribution to the development of pancreatic cancer remains unclear. This study showed that the expression of STC2 was significantly upregulated in pancreatic cancer tissues. Moreover, its expression was positively correlated with tumor size and lymph node metastasis and negatively correlated with 5-year survival rate of pancreatic cancer patients. Additionally, the expression levels of STC2 were a novel biomarker for predicting overall survival rate after surgery. Furthermore, overexpression of STC2 could promote the proliferation, migration, and invasion of pancreatic cancer cell lines, while knocking down of STC2 led to antiproliferation and antimetastasis activities. Further mechanistic investigations revealed that the expression of STC2 could significantly promote the epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. These data indicated that the overexpression of STC2 in pancreatic cancer contributes to the metastasis through the promotion of EMT, suggesting that STC2 is a potential prognostic biomarker and therapeutic target for pancreatic cancer.
Collapse
|
21
|
He H, Qie S, Guo Q, Chen S, Zou C, Lu T, Su Y, Zong J, Xu H, He D, Xu Y, Chen B, Pan J, Sang N, Lin S. Stanniocalcin 2 (STC2) expression promotes post-radiation survival, migration and invasion of nasopharyngeal carcinoma cells. Cancer Manag Res 2019; 11:6411-6424. [PMID: 31372045 PMCID: PMC6636319 DOI: 10.2147/cmar.s197607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 06/06/2019] [Indexed: 12/26/2022] Open
Abstract
Background: Stanniocalcin 2 (STC2) expression is upregulated under multiple stress conditions including hypoxia, nutrient starvation and radiation. Overexpression of STC2 correlates with tumor progression and poor prognosis. Purpose: We previously demonstrated that overexpression of STC2 in nasopharyngeal carcinomas (NPC) positively correlates with radiation resistance and tumor metastasis, two major clinical obstacles to the improvement of NPC management. However, it remains elusive whether STC2 expression is a critical contributing factor for post-radiation survival and metastasis of NPC cells. Materials and methods: Using the radiation resistant CNE2 cell line as a model, we examined the importance of STC2 expression for post-radiation survival, migration and invasion. Here, we report the establishment of STC2 knockout lines (CNE2-STC2-KO) using the CRISPR/Cas9-based genome editing technique. Results: Compared with the parental line, STC2-KO cells showed similar proliferation and morphology in normal culture conditions, and loss of STC2 did not compromise the cell tumorigenicity in nude mice model. However, STC2-KO lines demonstrated increased sensitivity to X-radiation under either normoxic or hypoxic conditions. Particularly, upon X-radiation, parental CNE2 cells only slightly whereas STC2-KO cells remarkably decreased the migration and invasion ability. Cell cycle analysis revealed that loss of STC2 accumulated cells in G1 and G2/M phases but decreased S-population. Conclusion: These data indicate that the expression of STC2, which can be stimulated by metabolic or therapeutic stresses, is one important factor to promote survival and metastasis of post-radiation NPC cells. Therefore, targeting STC2 or relative downstream pathways may provide novel strategies to overcome radiation resistance and metastasis of NPC.
Collapse
Affiliation(s)
- Huocong He
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Shuo Qie
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA.,Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Qiaojuan Guo
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Shuyang Chen
- Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Changyan Zou
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Tianzhu Lu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Ying Su
- Department of Radiation Biology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jingfeng Zong
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Hanchuan Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Dan He
- Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA
| | - Yun Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Bijuan Chen
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Jianji Pan
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| | - Nianli Sang
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA 19104, USA.,Department of Biology, Drexel University College of Arts & Sciences, Philadelphia, PA 19104, USA
| | - Shaojun Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University, Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
22
|
Li T, Qin Y, Zhen Z, Shen H, Cong T, Schiferle E, Xiao S. Long non-coding RNA HOTAIR/microRNA-206 sponge regulates STC2 and further influences cell biological functions in head and neck squamous cell carcinoma. Cell Prolif 2019; 52:e12651. [PMID: 31297902 PMCID: PMC6797510 DOI: 10.1111/cpr.12651] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 12/23/2022] Open
Abstract
Objective It is essential to characterize underlying molecular mechanism associated with head and neck squamous cell carcinoma (HNSCC) and identify promising therapeutic targets. Herein, we explored role of homeobox transcript antisense RNA (HOTAIR) in HNSCC to regulate stanniocalcin‐2 (STC2) by sponging microRNA‐206 (miR‐206). Methods HNSCC‐related differentially expressed genes and regulation network amongst HOTAIR, miR‐206 and STC2 were identified. Next, effect of HOTAIR on cell biological functions of HNSCC was identified after transfection of cells with HOTAIR overexpressed plasmids or siRNA against HOTAIR. PI3K/AKT signalling pathway‐related gene expression was measured after miR‐206 and STC2 were suppressed. Cell invasion, migration and proliferation were assessed. Finally, tumour growth was assessed to determine the effects of HOTAIR/miR‐206/STC2 axis in vivo. Results HOTAIR specifically bound to miR‐206 and miR‐206 targeted STC2. Downregulated HOTAIR or upregulated miR‐206 suppressed HNSCC cell proliferation, invasion and migration. miR‐206 inhibited PI3K/AKT signalling pathway by down‐regulating STC2. Besides, silenced HOTAIR or overexpressed miR‐206 repressed the tumour growth of nude mice with HNSCC. Conclusion HOTAIR regulated HNSCC cell biological functions by binding to miR‐206 through STC2.
Collapse
Affiliation(s)
- Tiancheng Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Yao Qin
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Zhen Zhen
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Hong Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Tiechuan Cong
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Erik Schiferle
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuifang Xiao
- Department of Otorhinolaryngology-Head and Neck Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
23
|
Liu Y, Tsai M, Wu S, Chang T, Tsai T, Gow C, Chang Y, Shih J. Acquired resistance to EGFR tyrosine kinase inhibitors is mediated by the reactivation of STC2/JUN/AXL signaling in lung cancer. Int J Cancer 2019; 145:1609-1624. [DOI: 10.1002/ijc.32487] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/06/2019] [Accepted: 05/21/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Yi‐Nan Liu
- Department of Internal MedicineNational Taiwan University Hospital Taipei Taiwan
| | - Meng‐Feng Tsai
- Department of Molecular BiotechnologyDa‐Yeh University Changhua Taiwan
| | - Shang‐Gin Wu
- Department of Internal MedicineNational Taiwan University Hospital Taipei Taiwan
- Department of Internal MedicineNational Taiwan University Cancer Center Taipei Taiwan
| | - Tzu‐Hua Chang
- Department of Internal MedicineNational Taiwan University Hospital Taipei Taiwan
| | - Tzu‐Hsiu Tsai
- Department of Internal MedicineNational Taiwan University Hospital Taipei Taiwan
| | - Chien‐Hung Gow
- Department of Internal MedicineFar Eastern Memorial Hospital New Taipei City Taiwan
| | - Yih‐Leong Chang
- Department of PathologyNational Taiwan University Hospital Taipei Taiwan
- Graduate Institute of Pathology, College of MedicineNational Taiwan University Taipei Taiwan
| | - Jin‐Yuan Shih
- Department of Internal MedicineNational Taiwan University Hospital Taipei Taiwan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan University Taipei Taiwan
| |
Collapse
|
24
|
Li JB, Liu ZX, Zhang R, Ma SP, Lin T, Li YX, Yang SH, Zhang WC, Wang YP. Sp1 contributes to overexpression of stanniocalcin 2 through regulation of promoter activity in colon adenocarcinoma. World J Gastroenterol 2019; 25:2776-2787. [PMID: 31236000 PMCID: PMC6580349 DOI: 10.3748/wjg.v25.i22.2776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/22/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Aberrant expression of stanniocalcin 2 (STC2) is implicated in colon adenocarcinoma (COAD). A previous study identified that STC2 functions as a tumor promoter to drive development of some cancers, but the role of its overexpression in the development of COAD remains unclear. AIM To evaluate the regulation mechanism of STC2 overexpression in COAD. METHODS The expression of STC2 in COAD was assessed by TCGA COAD database and GEO (GSE50760). Methylation level of the STC2 promoter was evaluated with beta value in UALCAN platform, and the correlation between STC2 expression and survival rate was investigated with TCGA COAD. Transcription binding site prediction was conducted by TRANSFAC and LASAGNA, and a luciferase reporter system was used to identify STC2 promoter activity in several cell lines, including HEK293T, NCM460, HT29, SW480, and HCT116. Western blotting was performed to evaluate the role of Sp1 on the expression of STC2. RESULTS The central finding of this work is that STC2 is overexpressed in COAD tissues and positively correlated with poor prognosis. Importantly, the binding site of the transcription factor Sp1 is widely located in the promoter region of STC2. A luciferase reporter system was successfully constructed to analyze the transcription activity of STC2, and knocking down the expression of Sp1 significantly inhibited the transcription activity of STC2. Furthermore, inhibition of Sp1 remarkably decreased protein levels of STC2. CONCLUSION Our data provide evidence that the transcription factor Sp1 is essential for the overexpression of STC2 in COAD through activation of promoter activity. Taken together, our finding provides new insights into the mechanism of oncogenic function of COAD by STC2.
Collapse
Affiliation(s)
- Ji-Bin Li
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Zhe-Xian Liu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Si-Ping Ma
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Tao Lin
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Yan-Xi Li
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| | - Shi-Hua Yang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
- China Medical University, Shenyang 110000, Liaoning Province, China
| | - Wan-Chuan Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
- China Medical University, Shenyang 110000, Liaoning Province, China
| | - Yong-Peng Wang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
25
|
Cockrum R, Speidel S, Crawford N, Zeng X, Blackburn H, Holt T, Enns R, Thomas M. Genotypes identified by genome-wide association analyses influence yearling pulmonary arterial pressure and growth traits in Angus heifers from a high-altitude beef production system. Livest Sci 2019. [DOI: 10.1016/j.livsci.2019.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
26
|
Sun G, Liu M, Han H. Overexpression of microRNA‐190 inhibits migration, invasion, epithelial‐mesenchymal transition, and angiogenesis through suppression of protein kinase B‐extracellular signal‐regulated kinase signaling pathway via binding to stanniocalicin 2 in breast cancer. J Cell Physiol 2019; 234:17824-17838. [PMID: 30993707 DOI: 10.1002/jcp.28409] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Guiming Sun
- Department of Oncology Liaocheng People's Hospital Liaocheng P.R. China
| | - Meirong Liu
- Department of Oncology Liaocheng People's Hospital Liaocheng P.R. China
| | - Hui Han
- Department of Oncology Liaocheng People's Hospital Liaocheng P.R. China
| |
Collapse
|
27
|
Zhang C, Chen S, Ma X, Yang Q, Su F, Shu X, Xie W, Feng M, Xiong B. Upregulation of STC2 in colorectal cancer and its clinicopathological significance. Onco Targets Ther 2019; 12:1249-1258. [PMID: 30863092 PMCID: PMC6389002 DOI: 10.2147/ott.s191609] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Stanniocalcin 2 (STC2) is a glycoprotein hormone involved in many biological processes and a secretory protein that regulates malignant tumor progression. The aim of the present study was to further explore the clinicopathological significance and prognostic role of STC2 in colorectal cancer (CRC). Methods In this study, STC2 expression was first investigated in Gene Expression Omnibus and The Cancer Genome Atlas, and then validated with the data from our medical center. Univariate and multivariate analyses were performed to assess the association between prognostic factors and survival outcome. Results In Gene Expression Omnibus and The Cancer Genome Atlas databases, bioinformatics analysis confirmed that STC2 was significantly increased in CRC compared with that in normal tissues (P<0.01), and CRC patients with high STC2 expression had a shorter overall survival. By analyzing data from our medical center, the results also showed that STC2 expression of CRC tissues was higher than that in normal tissues, whether the transcriptional or protein levels. In the CRC tissues, high STC2 expression was significantly correlated with lymph node metastasis (P=0.047), distant metastasis (P=0.040), and advanced clinical stage (P=0.047). Moreover, Kaplan–Meier analyses indicated that high STC2 expression predicted a worse prognosis, and multivariate Cox regression analysis revealed that STC2 was an independent prognostic factor for overall survival (HR =1.976, 95% CI: 1.092–3.576, P=0.024) in patients with CRC. Conclusion Our results suggested that STC2 played an important role in CRC progression and prognosis, and could be a useful biomarker for survival prediction.
Collapse
Affiliation(s)
- Chunxiao Zhang
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Shuangqian Chen
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Xiang Ma
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Qian Yang
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Fei Su
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Xiang Shu
- Department of Technology, Wuhan Hesheng Medical Technological Company, Wuhan 430071, China
| | - Wei Xie
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Maohui Feng
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| | - Bin Xiong
- Department of Gastrointestinal Surgery and Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Hubei Key Laboratoryof Tumor Biological Behaviors and Hubei Cancer Clinical Study Center, Wuchang District, Wuhan 430071, China, ;
| |
Collapse
|
28
|
Coulson-Gilmer C, Humphries MP, Sundara Rajan S, Droop A, Jackson S, Condon A, Cserni G, Jordan LB, Jones LJ, Kanthan R, Di Benedetto A, Mottolese M, Provenzano E, Kulka J, Shaaban AM, Hanby AM, Speirs V. Stanniocalcin 2 expression is associated with a favourable outcome in male breast cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2018; 4:241-249. [PMID: 29956502 PMCID: PMC6174618 DOI: 10.1002/cjp2.106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 11/11/2022]
Abstract
Breast cancer can occur in either gender; however, it is rare in men, accounting for <1% of diagnosed cases. In a previous transcriptomic screen of male breast cancer (MBC) and female breast cancer (FBC) occurrences, we observed that Stanniocalcin 2 (STC2) was overexpressed in the former. The aim of this study was to confirm the expression of STC2 in MBC and to investigate whether this had an impact on patient prognosis. Following an earlier transcriptomic screen, STC2 gene expression was confirmed by RT‐qPCR in matched MBC and FBC samples as well as in tumour‐associated fibroblasts derived from each gender. Subsequently, STC2 protein expression was examined immunohistochemically in tissue microarrays containing 477 MBC cases. Cumulative survival probabilities were calculated using the Kaplan–Meier method and multivariate survival analysis was performed using the Cox hazard model. Gender‐specific STC2 gene expression showed a 5.6‐fold upregulation of STC2 transcripts in MBC, also supported by data deposited in Oncomine™. STC2 protein expression was a positive prognostic factor for disease‐free survival (DFS; Log‐rank; total p = 0.035, HR = 0.49; tumour cells p = 0.017, HR = 0.44; stroma p = 0.030, HR = 0.48) but had no significant impact on overall survival (Log‐rank; total p = 0.23, HR = 0.71; tumour cells p = 0.069, HR = 0.59; stroma p = 0.650, HR = 0.87). Importantly, multivariate analysis adjusted for patient age at diagnosis, node staging, tumour size, ER, and PR status revealed that total STC2 expression as well as expression in tumour cells was an independent prognostic factor for DFS (Cox regression; p = 0.018, HR = 0.983; p = 0.015, HR = 0.984, respectively). In conclusion, STC2 expression is abundant in MBC where it is an independent prognostic factor for DFS.
Collapse
Affiliation(s)
| | - Matthew P Humphries
- Centre for Cancer Research and Cell Biology, Queen's University, Belfast, UK
| | | | - Alastair Droop
- MRC Medical Bioinformatics Centre, University of Leeds, Leeds, UK
| | - Sharon Jackson
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Alexandra Condon
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Gabor Cserni
- Department of Pathology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | | | | | - Rani Kanthan
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Anna Di Benedetto
- Department of Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | | | - Elena Provenzano
- Department of Histopathology, Addenbrooke's Hospital, Cambridge, UK
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Abeer M Shaaban
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham and University of Birmingham, Birmingham, UK
| | - Andrew M Hanby
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| |
Collapse
|
29
|
Ueda H, Akiyama Y, Shimada S, Mogushi K, Serizawa M, Matsumura S, Mitsunori Y, Aihara A, Ban D, Ochiai T, Kudo A, Tanabe M, Tanaka S. Tumor suppressor functions of DAXX through histone H3.3/H3K9me3 pathway in pancreatic NETs. Endocr Relat Cancer 2018; 25:619-631. [PMID: 29599123 DOI: 10.1530/erc-17-0328] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Pancreatic neuroendocrine tumors (PanNETs) have considerable malignant potential. Frequent somatic mutations and loss of DAXX protein expression have been found in PanNETs. DAXX is known as a transcriptional repressor; however, molecular functions underlying DAXX loss remain unclear in PanNETs. We evaluated DAXX expression by immunohistochemistry in 44 PanNETs. DAXX-knockdown (KD) and -knockout (KO) PanNET cells were analyzed for in vitro and vivo The target genes were screened by microarray and chromatin immunoprecipitation (ChIP) assays for DAXX, histone H3.3 and H3K9me3 complex. In clinicopathological features, low DAXX expression was significantly correlated with nonfunctional tumors, higher Ki-67 index and WHO grade. Microarray and ChIP assays of DAXX-KD/KO identified 12 genes as the direct targets of DAXX transcriptional repressor. Among them, expression of five genes including STC2 was suppressed by DAXX/H3.3/H3K9me3 pathway. DAXX-KD/KO cells enhanced sphere forming activity, but its effect was suppressed by knockdown of STC2 In xenograft models, tumorigenicity and tumor vessel density were significantly increased in DAXX-KO cells with high expression of STC2. Clinically, higher recurrence rate was recognized in PanNETs with low expression of DAXX and high expression of STC2 than others (P = 0.018). Our data suggest that DAXX plays as a tumor suppressor and DAXX/H3.3 complex suppresses target genes by promoting H3K9me3 in PanNETs. Combination of DAXX loss and its target gene STC2 overexpression might be effective biomarkers and therapeutic candidates.
Collapse
Affiliation(s)
- Hiroki Ueda
- Department of Molecular OncologyGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshimitsu Akiyama
- Department of Molecular OncologyGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shu Shimada
- Department of Molecular OncologyGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kaoru Mogushi
- Department of Molecular OncologyGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Misaki Serizawa
- Department of Molecular OncologyGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Matsumura
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Mitsunori
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Arihiro Aihara
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Ochiai
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular OncologyGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic SurgeryGraduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
30
|
López E, Gómez-Gordo L, Cantonero C, Bermejo N, Pérez-Gómez J, Granados MP, Salido GM, Rosado Dionisio JA, Redondo Liberal PC. Stanniocalcin 2 Regulates Non-capacitative Ca 2+ Entry and Aggregation in Mouse Platelets. Front Physiol 2018; 9:266. [PMID: 29628897 PMCID: PMC5876523 DOI: 10.3389/fphys.2018.00266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Stanniocalcin 2 (STC2) is a fish protein that controls body Ca2+ and phosphate metabolism. STC2 has also been described in mammals, and as platelet function highly depends on both extracellular and intracellular Ca2+, we have explored its expression and function in these cells. STC2−/− mice exhibit shorter tail bleeding time than WT mice. Platelets from STC2-deficient mice showed enhanced aggregation, as well as enhanced Ca2+ mobilization in response to the physiological agonist thrombin (Thr) and the diacylglycerol analog, OAG, a selective activator of the non-capacitative Ca2+ entry channels. Interestingly, platelets from STC2−/− mice exhibit attenuated interaction between STIM1 and Orai1 in response to Thr, thus suggesting that STC2 is required for Thr-evoked STIM1-Orai1 interaction and the subsequent store-operated Ca2+ entry (SOCE). We have further assessed possible changes in the expression of the most relevant channels involved in non-capacitative Ca2+ entry in platelets. Then, protein expression of Orai3, TRPC3 and TRPC6 were evaluated by Western blotting, and the results revealed that while the expression of Orai3 was enhanced in the STC2-deficient mice, others like TRPC3 and TRPC6 remains almost unaltered. Summarizing, our results provide for the first time evidence for a role of STC2 in platelet physiology through the regulation of agonist-induced Ca2+ entry, which might be mediated by the regulation of Orai3 channel expression.
Collapse
Affiliation(s)
- Esther López
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - L Gómez-Gordo
- Department of Animal Medicine, Veterinary Faculty University of Extremadura, Cáceres, Spain
| | - Carlos Cantonero
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Nuria Bermejo
- Hematology Unit, San Pedro de Alcantara Hospital, Cáceres, Spain
| | - Jorge Pérez-Gómez
- Faculty of Sport Sciences, University of Extremadura, Cáceres, Spain
| | - María P Granados
- Aldea Moret Health Center, Extremadura Health Service, Cáceres, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, Cáceres, Spain
| | - Juan A Rosado Dionisio
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Pedro C Redondo Liberal
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| |
Collapse
|
31
|
Multiplicity of acquired cross-resistance in paclitaxel-resistant cancer cells is associated with feedback control of TUBB3 via FOXO3a-mediated ABCB1 regulation. Oncotarget 2018; 7:34395-419. [PMID: 27284014 PMCID: PMC5085164 DOI: 10.18632/oncotarget.9118] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/11/2016] [Indexed: 12/22/2022] Open
Abstract
Acquired drug resistance is a primary obstacle for effective cancer therapy. The correlation of point mutations in class III β-tubulin (TUBB3) and the prominent overexpression of ATP-binding cassette P-glycoprotein (ABCB1), a multidrug resistance gene, have been protruding mechanisms of resistance to microtubule disruptors such as paclitaxel (PTX) for many cancers. However, the precise underlying mechanism of the rapid onset of cross-resistance to an array of structurally and functionally unrelated drugs in PTX-resistant cancers has been poorly understood. We determined that our established PTX-resistant cancer cells display ABCB1/ABCC1-associated cross-resistance to chemically different drugs such as 5-fluorouracil, docetaxel, and cisplatin. We found that feedback activation of TUBB3 can be triggered through the FOXO3a-dependent regulation of ABCB1, which resulted in the accentuation of induced PTX resistance and encouraged multiplicity in acquired cross-resistance. FOXO3a-directed regulation of P-glycoprotein (P-gp) function suggests that control of ABCB1 involves methylation-dependent activation. Consistently, transcriptional overexpression or downregulation of FOXO3a directs inhibitor-controlled protease-degradation of TUBB3. The functional PI3K/Akt signaling is tightly responsive to FOXO3a activation alongside doxorubicin treatment, which directs FOXO3a arginine hypermethylation. In addition, we found that secretome factors from PTX-resistant cancer cells with acquired cross-resistance support a P-gp-dependent association in multidrug resistance (MDR) development, which assisted the FOXO3a-mediated control of TUBB3 feedback. The direct silencing of TUBB3 reverses induced multiple cross-resistance, reduces drug-resistant tumor mass, and suppresses the impaired microtubule stability status of PTX-resistant cells with transient cross-resistance. These findings highlight the control of the TUBB3 response to ABCB1 genetic suppressors as a mechanism to reverse the profuse development of multidrug resistance in cancer.
Collapse
|
32
|
López JJ, Jardín I, Cantonero Chamorro C, Duran ML, Tarancón Rubio MJ, Reyes Panadero M, Jiménez F, Montero R, González MJ, Martínez M, Hernández MJ, Brull JM, Corbacho AJ, Delgado E, Granados MP, Gómez-Gordo L, Rosado JA, Redondo PC. Involvement of stanniocalcins in the deregulation of glycaemia in obese mice and type 2 diabetic patients. J Cell Mol Med 2017; 22:684-694. [PMID: 28990324 PMCID: PMC5742690 DOI: 10.1111/jcmm.13355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/19/2017] [Indexed: 12/18/2022] Open
Abstract
Stanniocalcins are expressed in the pancreas tissue, and it was suggested a direct correlation between circulating insulin and STC2 concentrations in human. Here, we show a significant correlation between STC1 and both glycaemia and glycosylated haemoglobin among DM2 patients, while DM2 patients who present the greatest glycosylated haemoglobin values exhibited the lowest STC2 expression. However, treatment of patients with antiglycaemic drugs does not significantly modify the expression of both STCs. On the other hand, STC2‐/‐ mice that exhibited neonatal and adult overweight further presented deregulated glycaemia when they were feed with a hypercaloric diet (breeding pellet, BP). This alteration is more evident at the early stages of the animal life. Deregulated glycaemia in these mice was confirmed using glucose oral test. In addition, STC2‐/‐ mice present enhanced pancreas size; thus, the histological analysis reveals that WT mice respond to BP diet by increasing the size of the pancreatic islets through inducing cell division, and STC2‐/‐ mice lack this compensatory mechanism. Contrary, BP fed STC2‐/‐ mice show enhanced number of islets but of similar size than those fed with regular pellet. Histopathological analysis demonstrates tissue structure disruption and erythrocytes infiltrations in STC2‐/‐ mice, possibly due to the stress evoked by the BP diet. Finally, enhanced glucagon immunostaining was observed in the islet of STC2‐/‐ mice, and the glucagon ELISA assay confirmed the increase in the circulating glucagon. Summarizing, we present evidence of the role of STCs, mainly STC2, as a possible early marker during development of diabetes mellitus.
Collapse
Affiliation(s)
- José Javier López
- Department of Physiology (Phycell), Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Isaac Jardín
- Department of Physiology (Phycell), Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | | | - Manuel Luis Duran
- Animal House of University of Extremadura, University of Extremadura, Cáceres, Spain
| | | | - Maria Reyes Panadero
- Animal House of University of Extremadura, University of Extremadura, Cáceres, Spain
| | - Francisca Jiménez
- Manuel Encinas's medical center, Extremadura Health Service, Cáceres, Spain
| | - Rocio Montero
- Manuel Encinas's medical center, Extremadura Health Service, Cáceres, Spain
| | | | - Manuel Martínez
- Manuel Encinas's medical center, Extremadura Health Service, Cáceres, Spain
| | | | | | | | | | | | - Luis Gómez-Gordo
- Department of Animal Medicine, Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Juan Antonio Rosado
- Department of Physiology (Phycell), Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Pedro Cosme Redondo
- Department of Physiology (Phycell), Veterinary Faculty, University of Extremadura, Cáceres, Spain
| |
Collapse
|
33
|
Huang R, Chen Z, He L, He N, Xi Z, Li Z, Deng Y, Zeng X. Mass spectrometry-assisted gel-based proteomics in cancer biomarker discovery: approaches and application. Theranostics 2017; 7:3559-3572. [PMID: 28912895 PMCID: PMC5596443 DOI: 10.7150/thno.20797] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022] Open
Abstract
There is a critical need for the discovery of novel biomarkers for early detection and targeted therapy of cancer, a major cause of deaths worldwide. In this respect, proteomic technologies, such as mass spectrometry (MS), enable the identification of pathologically significant proteins in various types of samples. MS is capable of high-throughput profiling of complex biological samples including blood, tissues, urine, milk, and cells. MS-assisted proteomics has contributed to the development of cancer biomarkers that may form the foundation for new clinical tests. It can also aid in elucidating the molecular mechanisms underlying cancer. In this review, we discuss MS principles and instrumentation as well as approaches in MS-based proteomics, which have been employed in the development of potential biomarkers. Furthermore, the challenges in validation of MS biomarkers for their use in clinical practice are also reviewed.
Collapse
Affiliation(s)
- Rongrong Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhongsi Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lei He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Nongyue He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Economical Forest Cultivation and Utilization of 2011 Collaborative Innovation Center in Hunan Province, Hunan Key Laboratory of Green Chemistry and Application of Biological Nanotechnology; Hunan University of Technology, Zhuzhou 412007, China
| | - Zhijiang Xi
- School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhiyang Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Department of Clinical Laboratory, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yan Deng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Economical Forest Cultivation and Utilization of 2011 Collaborative Innovation Center in Hunan Province, Hunan Key Laboratory of Green Chemistry and Application of Biological Nanotechnology; Hunan University of Technology, Zhuzhou 412007, China
| | - Xin Zeng
- Nanjing Maternity and Child Health Medical Institute, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| |
Collapse
|
34
|
Aldonza MBD, Son YS, Sung HJ, Ahn JM, Choi YJ, Kim YI, Cho S, Cho JY. Paraoxonase-1 (PON1) induces metastatic potential and apoptosis escape via its antioxidative function in lung cancer cells. Oncotarget 2017; 8:42817-42835. [PMID: 28467805 PMCID: PMC5522108 DOI: 10.18632/oncotarget.17069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/20/2017] [Indexed: 01/19/2023] Open
Abstract
Paraoxonase-1 (PON1) gene polymorphisms have been closely associated with the development of advanced cancers while PON1 secretion to the serum is linked with inhibition of oxidized high-density lipoprotein by its antioxidative function. Our group previously demonstrated that post-translational modification of serum PON1 in form of fucosylated PON1 is a potential biomarker of small cell lung cancer. Here, we interrogated the role of PON1 in the pathobiology of lung cancer (LC) by addressing cell-autonomous mechanisms using gain-of-function and loss-of-function approaches and protein expression profiling of tissue samples in our clinical biobank. PON1 expression in LC patient tissues varied between overexpression in squamous cell carcinoma and minimal loss in adenocarcinoma sub-types. Simultaneous overexpression of PON1 both at the gene and protein stability levels induced pro-oncogenic characteristics in LC cells and xenografts. PON1 overexpression supported metastatic progression of LC by decreasing G1/S ratio and LC cell senescence involving p21Waf1/Cip1. PON1 suppressed drug- and ligand-induced cell death and protected LC cells from genotoxic damages with maintained ATP levels, requiring p53-directed signals. PON1 promoted ROS deregulation protecting the mitochondria from dysregulation. PON1 knockdown resulted in the blockage of its antioxidant function in LC cells through Akt signaling with reduced invasive signature as a consequence of scant expression. Targeted glycolysis stimulated PON1 antioxidant activity regulating phosphorylation of AMPK-α. The functional data imply that exploitation of the antioxidative function of PON1 is consequential in driving LC pathogenesis at the cell-autonomous mechanistic level with consequences on tumor growth.
Collapse
Affiliation(s)
- Mark Borris D. Aldonza
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Current address: Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Yeon Sung Son
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hye-Jin Sung
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jung Mo Ahn
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Current address: Bio Center, Incheon Technopark, Incheon, Republic of Korea
| | - Young-Jin Choi
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Current address: College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Yong-In Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoungnam-Si, Gyeonggi-Do, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Aldonza MBD, Hong JY, Lee SK. Paclitaxel-resistant cancer cell-derived secretomes elicit ABCB1-associated docetaxel cross-resistance and escape from apoptosis through FOXO3a-driven glycolytic regulation. Exp Mol Med 2017; 49:e286. [PMID: 28104912 PMCID: PMC5291837 DOI: 10.1038/emm.2016.131] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 01/12/2023] Open
Abstract
Chemotherapy-induced cancer cell secretomes promote resistance due, in part, to a predominant glycolytic energy metabolism, which drives aggressive cancer cell proliferation. However, the characterization of these secretomes and the molecular events that associate them with acquired drug resistance remain poorly understood. In this study, we show that secretomes of cancer cells with high-level paclitaxel resistance stimulated cell proliferation and suppressed drug-induced apoptosis of drug-sensitive cells. We also found that drug (docetaxel)-stimulated induction of interferon-α (IFN-α), IFN-λ and tumor necrosis factor-α (TNF-α) release in drug-sensitive cells was lowered by these secretomes. The promotion of cell proliferation by paclitaxel-resistant (PacR) cancer cell secretomes was associated, in part, with an increase in S phase of the cell cycle and downregulation of the cell death pathway that supports escape from apoptosis. In addition, we also found that the regulation of targeted glycolysis in PacR cancer cells alters the effects of the secretomes on cell growth, apoptosis, ATP generation and acquired drug resistance. Further study revealed that the deletion of FOXO3a transcription exacerbates glycolytic shift-induced apoptosis by rescuing TRAIL expression. By generating a docetaxel-cross-resistant PacR cancer cell line (PacR/DCT), we further clarified the role of FOXO3a in glycolysis-associated mediation of P-glycoprotein/ABCB1 hyperactivity that induces docetaxel cross-resistance. These findings suggest that suppression of the cellular energy supply by targeting glycolysis may inhibit the multiplicity of acquired chemotherapy resistance. Therefore, the therapeutic inhibition of FOXO3a might direct glycolysis to induce apoptosis and overcome multidrug resistance in cancer cells.
Collapse
Affiliation(s)
- Mark Borris D Aldonza
- College of Pharmacy, Seoul National University, Seoul, Korea.,Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Ji-Young Hong
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
36
|
Pacheco J, Vaca L. STIM-TRP Pathways and Microdomain Organization: Auxiliary Proteins of the STIM/Orai Complex. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:189-210. [DOI: 10.1007/978-3-319-57732-6_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
37
|
Bosse K, Haneder S, Arlt C, Ihling CH, Seufferlein T, Sinz A. Mass spectrometry-based secretome analysis of non-small cell lung cancer cell lines. Proteomics 2016; 16:2801-2814. [DOI: 10.1002/pmic.201600297] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/24/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Konstanze Bosse
- Department of Pharmaceutical Chemistry & Bioanalytics; Institute of Pharmacy; Martin-Luther University Halle-Wittenberg; Halle (Saale) Germany
| | | | - Christian Arlt
- Department of Pharmaceutical Chemistry & Bioanalytics; Institute of Pharmacy; Martin-Luther University Halle-Wittenberg; Halle (Saale) Germany
| | - Christian H. Ihling
- Department of Pharmaceutical Chemistry & Bioanalytics; Institute of Pharmacy; Martin-Luther University Halle-Wittenberg; Halle (Saale) Germany
| | | | - Andrea Sinz
- Department of Pharmaceutical Chemistry & Bioanalytics; Institute of Pharmacy; Martin-Luther University Halle-Wittenberg; Halle (Saale) Germany
| |
Collapse
|
38
|
Fjeldbo CS, Aarnes EK, Malinen E, Kristensen GB, Lyng H. Identification and Validation of Reference Genes for RT-qPCR Studies of Hypoxia in Squamous Cervical Cancer Patients. PLoS One 2016; 11:e0156259. [PMID: 27244197 PMCID: PMC4887009 DOI: 10.1371/journal.pone.0156259] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 05/11/2016] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an adverse factor in cervical cancer, and hypoxia-related gene expression could be a powerful biomarker for identifying the aggressive hypoxic tumors. Reverse transcription quantitative PCR (RT-qPCR) is a valuable method for gene expression studies, but suitable reference genes for data normalization that are independent of hypoxia status and clinical parameters of cervical tumors are lacking. In the present work, we aimed to identify reference genes for RT-qPCR studies of hypoxia in squamous cervical cancer. From 422 candidate reference genes selected from the literature, we used Illumina array-based expression profiles to identify 182 genes not affected by hypoxia in cervical cancer, i.e. genes regulated by hypoxia in eight cervical cancer cell lines or correlating with the hypoxia-associated dynamic contrast-enhanced magnetic resonance imaging parameter ABrix in 42 patients, were excluded. Among the 182 genes, nine candidates (CHCHD1, GNB2L1, IPO8, LASP1, RPL27A, RPS12, SOD1, SRSF9, TMBIM6) that were not associated with tumor volume, stage, lymph node involvement or disease progression in array data of 150 patients, were selected for further testing by RT-qPCR. geNorm and NormFinder analyses of RT-qPCR data of 74 patients identified CHCHD1, SRSF9 and TMBIM6 as the optimal set of reference genes, with stable expression both overall and across patient subgroups with different hypoxia status (ABrix) and clinical parameters. The suitability of the three reference genes were validated in studies of the hypoxia-induced genes DDIT3, ERO1A, and STC2. After normalization, the RT-qPCR data of these genes showed a significant correlation with Illumina expression (P<0.001, n = 74) and ABrix (P<0.05, n = 32), and the STC2 data were associated with clinical outcome, in accordance with the Illumina data. Thus, CHCHD1, SRSF9 and TMBIM6 seem to be suitable reference genes for studying hypoxia-related gene expression in squamous cervical cancer samples by RT-qPCR. Moreover, STC2 is a promising prognostic hypoxia biomarker in cervical cancer.
Collapse
Affiliation(s)
- Christina S. Fjeldbo
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Eva-Katrine Aarnes
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Medical Physics, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Gunnar B. Kristensen
- Department of Gynaecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Heidi Lyng
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- * E-mail:
| |
Collapse
|
39
|
Qiu F, Yang L, Lu X, Chen J, Wu D, Wei Y, Nong Q, Zhang L, Fang W, Chen X, Ling X, Yang B, Zhang X, Zhou Y, Lu J. The MKK7 p.Glu116Lys Rare Variant Serves as a Predictor for Lung Cancer Risk and Prognosis in Chinese. PLoS Genet 2016; 12:e1005955. [PMID: 27028764 PMCID: PMC4814107 DOI: 10.1371/journal.pgen.1005955] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/03/2016] [Indexed: 11/19/2022] Open
Abstract
Accumulated evidence indicates that rare variants exert a vital role on predisposition and progression of human diseases, which provides neoteric insights into disease etiology. In the current study, based on three independently retrospective studies of 5,016 lung cancer patients and 5,181 controls, we analyzed the associations between five rare polymorphisms (i.e., p.Glu116Lys, p.Asn118Ser, p.Arg138Cys, p.Ala195Thr and p.Leu259Phe) in MKK7 and lung cancer risk and prognosis. To decipher the precise mechanisms of MKK7 rare variants on lung cancer, a series of biological experiments was further performed. We found that the MKK7 p.Glu116Lys rare polymorphism was significantly associated with lung cancer risk, progression and prognosis. Compared with Glu/Glu common genotype, the 116Lys rare variants (Lys/Glu/+ Lys/Lys) presented an adverse effect on lung cancer susceptibility (odds ratio [OR] = 3.29, 95% confidence interval [CI] = 2.70-4.01). These rare variants strengthened patients' clinical progression that patients with 116Lys variants had a significantly higher metastasis rate and advanced N, M stages at diagnosis. In addition, the patients with 116Lys variants also contributed to worse cancer prognosis than those carriers with Glu/Glu genotype (hazard ratio [HR] = 1.53, 95% CI = 1.32-1.78). Functional experiments further verified that the MKK7 p.116Lys variants altered the expression of several cancer-related genes and thus affected lung cancer cells proliferation, tumor growth and metastasis in vivo and in vitro. Taken together, our findings proposed that the MKK7 p.Glu116Lys rare polymorphism incurred a pernicious impact on lung cancer risk and prognosis through modulating expressions of a serial of cancer-related genes.
Collapse
Affiliation(s)
- Fuman Qiu
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
- Biomedicine Research Center and Department of Surgery, The Third Affiliated Hospital of Guangzhou Medicine University, Guangzhou, People's Republic of China
| | - Lei Yang
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoxiao Lu
- School of Arts and Sciences, Colby-Sawyer College, New London, New Hampshire, United States of America
| | - Jiansong Chen
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Di Wu
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yongfang Wei
- Center of Laboratory Animal, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qingqing Nong
- Department of Environmental Health, Guangxi Medical University, Nanning, People's Republic of China
| | - Lisha Zhang
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Wenxiang Fang
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoliang Chen
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaoxuan Ling
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Binyao Yang
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yifeng Zhou
- Department of Genetics, Medical College of Soochow University, Suzhou, People's Republic of China
| | - Jiachun Lu
- The State Key Lab of Respiratory Disease, The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
40
|
Bagordakis E, Sawazaki-Calone I, Macedo CCS, Carnielli CM, de Oliveira CE, Rodrigues PC, Rangel ALCA, Dos Santos JN, Risteli J, Graner E, Salo T, Paes Leme AF, Coletta RD. Secretome profiling of oral squamous cell carcinoma-associated fibroblasts reveals organization and disassembly of extracellular matrix and collagen metabolic process signatures. Tumour Biol 2016; 37:9045-57. [PMID: 26762409 DOI: 10.1007/s13277-015-4629-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/10/2015] [Indexed: 01/23/2023] Open
Abstract
An important role has been attributed to cancer-associated fibroblasts (CAFs) in the tumorigenesis of oral squamous cell carcinoma (OSCC), the most common tumor of the oral cavity. Previous studies demonstrated that CAF-secreted molecules promote the proliferation and invasion of OSCC cells, inducing a more aggressive phenotype. In this study, we searched for differences in the secretome of CAFs and normal oral fibroblasts (NOF) using mass spectrometry-based proteomics and biological network analysis. Comparison of the secretome profiles revealed that upregulated proteins involved mainly in extracellular matrix organization and disassembly and collagen metabolism. Among the upregulated proteins were fibronectin type III domain-containing 1 (FNDC1), serpin peptidase inhibitor type 1 (SERPINE1), and stanniocalcin 2 (STC2), the upregulation of which was validated by quantitative PCR and ELISA in an independent set of CAF cell lines. The transition of transforming growth factor beta 1 (TGF-β1)-mediating NOFs into CAFs was accompanied by significant upregulation of FNDC1, SERPINE1, and STC2, confirming the participation of these proteins in the CAF-derived secretome. Type I collagen, the main constituent of the connective tissue, was also associated with several upregulated biological processes. The immunoexpression of type I collagen N-terminal propeptide (PINP) was significantly correlated in vivo with CAFs in the tumor front and was associated with significantly shortened survival of OSCC patients. Presence of CAFs in the tumor stroma was also an independent prognostic factor for OSCC disease-free survival. These results demonstrate the value of secretome profiling for evaluating the role of CAFs in the tumor microenvironment and identify potential novel therapeutic targets such as FNDC1, SERPINE1, and STC2. Furthermore, type I collagen expression by CAFs, represented by PINP levels, may be a prognostic marker of OSCC outcome.
Collapse
Affiliation(s)
- Elizabete Bagordakis
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Iris Sawazaki-Calone
- Oral Pathology and Oral Medicine, Dentistry School, Western Paraná State University, Cascavel, PR, Brazil
| | - Carolina Carneiro Soares Macedo
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Carolina M Carnielli
- Brazilian Biociences National Laboratory-CNPEM, CEP 13083-970, Campinas, SP, Brazil
| | - Carine Ervolino de Oliveira
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Priscila Campioni Rodrigues
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Ana Lucia C A Rangel
- Oral Pathology and Oral Medicine, Dentistry School, Western Paraná State University, Cascavel, PR, Brazil
| | - Jean Nunes Dos Santos
- Laboratory of Surgical Pathology, Dental School, Federal University of Bahia-UFBA, Salvador, BA, Brazil
| | - Juha Risteli
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Edgard Graner
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil
| | - Tuula Salo
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Medical Research Center, Oulu University Hospital, Oulu, Finland.,Oral and Maxillofacial Diseases Unit, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| | | | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, State University of Campinas, Av. Limeira 901, CEP 13414-018, Piracicaba, SP, Brazil.
| |
Collapse
|