1
|
Vienot A, Vernerey D, Bouard A, Klajer E, Kim S, Tournigand C, Louvet C, André T, Rousseau B, Wespiser M, Spehner L, Wang YA, Weispfenning A, Dochy E, Borg C. Stanniocalcin 1 in Patients with Refractory Colorectal Cancer Treated with Regorafenib: A Post Hoc Biomarker Analysis of the TEXCAN and CORRECT Trials. CANCER RESEARCH COMMUNICATIONS 2025; 5:287-294. [PMID: 39807836 PMCID: PMC11811826 DOI: 10.1158/2767-9764.crc-24-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 09/27/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
SIGNIFICANCE STC1 is a protein secreted by intratumor endothelial cells in which plasma concentrations increase in patients with chemorefractory mCRC. Based on analyses of patients with refractory mCRC in the TEXCAN and CORRECT trials, we found that STC1 plasma levels had a prognostic role for OS, with high levels associated with poor outcome. A predictive role for baseline STC1 levels was pointed out for regorafenib efficacy.
Collapse
Affiliation(s)
- Angélique Vienot
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Dewi Vernerey
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Adeline Bouard
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Elodie Klajer
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Stefano Kim
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | | | | | - Thierry André
- Sorbonne Université and Hôpital Saint-Antoine, Paris, France
| | | | - Mylène Wespiser
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Laurie Spehner
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| | - Ying A. Wang
- Bayer HealthCare Pharmaceuticals, Cambridge, Massachusetts
| | | | | | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
- University of Franche Comté, EFS, INSERM, UMR RIGHT, Besançon, France
| |
Collapse
|
2
|
Wang K, Liu Y, Li S, Zhao N, Qin F, Tao Y, Song Z. Unveiling the therapeutic potential and mechanisms of stanniocalcin-1 in retinal degeneration. Surv Ophthalmol 2025; 70:106-120. [PMID: 39270826 DOI: 10.1016/j.survophthal.2024.08.001] [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: 11/29/2023] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024]
Abstract
Retinal degeneration (RD) is a group of ocular diseases characterized by progressive photoreceptor apoptosis and visual impairment. Mitochondrial malfunction, excessive oxidative stress, and chronic activation of neuroglia collectively contribute to the development of RD. Currently, there is a lack of efficacious therapeutic interventions for RD. Stanniocalcin-1 (STC-1) is a promising candidate molecule to decelerate photoreceptor cell death. STC-1 is a secreted calcium/phosphorus regulatory protein that exerts diverse protective effects. Accumulating evidence suggests that STC-1 protects retinal cells from ischemic injury, oxidative stress, and excessive apoptosis through enhancing the expression of uncoupling protein-2 (UCP-2). Furthermore, STC-1 exerts its antiinflammatory effects by inhibiting the activation of microglia and macrophages, as well as the synthesis and secretion of proinflammatory cytokines, such as TNF-α, IL-1, and IL-6. By employing these mechanisms, STC-1 effectively shields the retinal photoreceptors and optic nerve, thereby slowing down the progression of RD. We summarize the STC-1-mediated therapeutic effects on the degenerating retina, with a particular focus on its underlying mechanisms. These findings highlight that STC-1 may act as a versatile molecule to treat degenerative retinopathy. Further research on STC-1 is imperative to establish optimal protocols for its clinical use.
Collapse
Affiliation(s)
- Kexin Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Yashuang Liu
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Na Zhao
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China
| | - Fangyuan Qin
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Ye Tao
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| | - Zongming Song
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| |
Collapse
|
3
|
Alkafaas SS, Khedr SA, ElKafas SS, Hafez W, Loutfy SA, Sakran M, Janković N. Targeting JNK kinase inhibitors via molecular docking: A promising strategy to address tumorigenesis and drug resistance. Bioorg Chem 2024; 153:107776. [PMID: 39276490 DOI: 10.1016/j.bioorg.2024.107776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/13/2024] [Accepted: 08/28/2024] [Indexed: 09/17/2024]
Abstract
Among members of the mitogen-activated protein kinase (MAPK) family, c-Jun N-terminal kinases (JNKs) are vital for cellular responses to stress, inflammation, and apoptosis. Recent advances have highlighted their important implications in cancer biology, where dysregulated JNK signalling plays a role in the growth, progression, and metastasis of tumors. The present understanding of JNK kinase and its function in the etiology of cancer is summarized in this review. By modifying a number of downstream targets, such as transcription factors, apoptotic regulators, and cell cycle proteins, JNKs exert diverse effects on cancer cells. Apoptosis avoidance, cell survival, and proliferation are all promoted by abnormal JNK activation in many types of cancer, which leads to tumor growth and resistance to treatment. JNKs also affect the tumour microenvironment by controlling the generation of inflammatory cytokines, angiogenesis, and immune cell activity. However, challenges remain in deciphering the context-specific roles of JNK isoforms and their intricate crosstalk with other signalling pathways within the complex tumor environment. Further research is warranted to delineate the precise mechanisms underlying JNK-mediated tumorigenesis and to develop tailored therapeutic strategies targeting JNK signalling to improve cancer management. The review emphasizes the role of JNK kinases in cancer biology, as well as their potential as pharmaceutical targets for precision oncology therapy and cancer resistance. Also, this review summarizes all the available promising JNK inhibitors that are suggested to promote the responsiveness of cancer cells to cancer treatment.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, 31527, Egypt.
| | - Sohila A Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Sara Samy ElKafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt; Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, Russia
| | - Wael Hafez
- NMC Royal Hospital, 16th St - Khalifa City - SE-4 - Abu Dhabi, United Arab Emirates; Department of Internal Medicine, Medical Research and Clinical Studies Institute, The National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, Cairo Governorate 12622, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mohamed Sakran
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Nenad Janković
- Institute for Information Technologies Kragujevac, Department of Science, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia.
| |
Collapse
|
4
|
Feng G, Yang X, Shuai W, Wang G, Ouyang L. Update on JNK inhibitor patents: 2015 to present. Expert Opin Ther Pat 2024; 34:907-927. [PMID: 39223788 DOI: 10.1080/13543776.2024.2400167] [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: 04/10/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION c-Jun N-terminal kinase (JNK) regulates various biological processes through the phosphorylation cascade and is closely associated with numerous diseases, including inflammation, cardiovascular diseases, and neurological disorders. Therefore, JNKs have emerged as potential targets for disease treatment. AREAS COVERED This review compiles the patents and literatures concerning JNK inhibitors through retrieving relevant information from the SciFinder, Google Patents databases, and PubMed from 2015 to the present. It summarizes the structure-activity relationship (SAR) and biological activity profiles of JNK inhibitors, offering valuable perspectives on their potential therapeutic applications. EXPERT OPINION The JNK kinase serves as a novel target for the treatment of neurodegenerative disorders, pulmonary fibrosis, and other illnesses. A variety of small-molecule inhibitors targeting JNKs have demonstrated promising therapeutic potential in preclinical studies, which act upon JNK kinases via distinct mechanisms, encompassing traditional ATP competitive inhibition, covalent inhibition, and bidentate inhibition. Among them, several JNK inhibitors from PregLem SA, Celegene SA, and Xigen SA have accomplished the early stage of clinical trials, and their results will guide the development and indications of future JNK inhibitors.
Collapse
Affiliation(s)
| | | | | | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and West China second Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and West China second Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Duan C, He B, Wang Y, Liu W, Bao W, Yu L, Xin J, Gui H, Lei J, Yang Z, Liu J, Tao W, Qin J, Luo J, Dong Z. Stanniocalcin-1 promotes temozolomide resistance of glioblastoma through regulation of MGMT. Sci Rep 2024; 14:20199. [PMID: 39215105 PMCID: PMC11364827 DOI: 10.1038/s41598-024-68902-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Temozolomide (TMZ) resistance is a major challenge in the treatment of glioblastoma (GBM). Tumour reproductive cells (TRCs) have been implicated in the development of chemotherapy resistance. By culturing DBTRG cells in three-dimensional soft fibrin gels to enrich GBM TRCs and performing RNA-seq analysis, the expression of stanniocalcin-1 (STC), a gene encoding a secreted glycoprotein, was found to be upregulated in TRCs. Meanwhile, the viability of TMZ-treated TRC cells was significantly higher than that of TMZ-treated 2D cells. Analysis of clinical data from CGGA (Chinese Glioma Genome Atlas) database showed that high expression of STC1 was closely associated with poor prognosis, glioma grade and resistance to TMZ treatment, suggesting that STC1 may be involved in TMZ drug resistance. The expression of STC1 in tissues and cells was examined, as well as the effect of STC1 on GBM cell proliferation and TMZ-induced DNA damage. The results showed that overexpression of STC1 promoted and knockdown of STC1 inhibited TMZ-induced DNA damage. These results were validated in an intracranial tumour model. These data revealed that STC1 exerts regulatory functions on MGMT expression in GBM, and provides a rationale for targeting STC1 to overcome TMZ resistance.
Collapse
Affiliation(s)
- Chao Duan
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Bincan He
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Yiqi Wang
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Wanying Liu
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Wendai Bao
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Li Yu
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Jinxin Xin
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Hui Gui
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Junrong Lei
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Zehao Yang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Jun Liu
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Weiwei Tao
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Jun Qin
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China.
| | - Jie Luo
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China.
| | - Zhiqiang Dong
- Center for Neurological Disease Research, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China.
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
- Central Laboratory, Hubei Cancer Hospital, Wuhan, 430070, Hubei, China.
| |
Collapse
|
6
|
Gali A, Bijnsdorp IV, Piersma SR, Pham TV, Gutiérrez-Galindo E, Kühnel F, Tsolakos N, Jimenez CR, Hausser A, Alexopoulos LG. Protein kinase D drives the secretion of invasion mediators in triple-negative breast cancer cell lines. iScience 2024; 27:108958. [PMID: 38323010 PMCID: PMC10844833 DOI: 10.1016/j.isci.2024.108958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/28/2023] [Accepted: 01/15/2024] [Indexed: 02/08/2024] Open
Abstract
The protein kinase D (PKD) family members regulate the fission of cargo vesicles at the Golgi complex and play a pro-oncogenic role in triple-negative breast cancer (TNBC). Whether PKD facilitates the secretion of tumor-promoting factors in TNBC, however, is still unknown. Using the pharmacological inhibition of PKD activity and siRNA-mediated depletion of PKD2 and PKD3, we identified the PKD-dependent secretome of the TNBC cell lines MDA-MB-231 and MDA-MB-468. Mass spectrometry-based proteomics and antibody-based assays revealed a significant downregulation of extracellular matrix related proteins and pro-invasive factors such as LIF, MMP-1, MMP-13, IL-11, M-CSF and GM-CSF in PKD-perturbed cells. Notably, secretion of these proteins in MDA-MB-231 cells was predominantly controlled by PKD2 and enhanced spheroid invasion. Consistently, PKD-dependent secretion of pro-invasive factors was more pronounced in metastatic TNBC cell lines. Our study thus uncovers a novel role of PKD2 in releasing a pro-invasive secretome.
Collapse
Affiliation(s)
- Alexia Gali
- Biomedical Systems Laboratory, National Technical University of Athens, 15780 Athens, Greece
- Protavio Ltd, Demokritos Science Park, 15341 Athens, Greece
| | - Irene V. Bijnsdorp
- Department of Urology, Cancer Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, de Boelelaan 1117, Amsterdam 1081 HV, the Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | - Sander R. Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | - Thang V. Pham
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | | | - Fiona Kühnel
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Nikos Tsolakos
- Protavio Ltd, Demokritos Science Park, 15341 Athens, Greece
| | - Connie R. Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, OncoProteomics Laboratory, de Boelelaan 1117, , Amsterdam 1081 HV, the Netherlands
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
- Stuttgart Research Center for Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Leonidas G. Alexopoulos
- Biomedical Systems Laboratory, National Technical University of Athens, 15780 Athens, Greece
- Protavio Ltd, Demokritos Science Park, 15341 Athens, Greece
| |
Collapse
|
7
|
Emerging phagocytosis checkpoints in cancer immunotherapy. Signal Transduct Target Ther 2023; 8:104. [PMID: 36882399 PMCID: PMC9990587 DOI: 10.1038/s41392-023-01365-z] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 03/09/2023] Open
Abstract
Cancer immunotherapy, mainly including immune checkpoints-targeted therapy and the adoptive transfer of engineered immune cells, has revolutionized the oncology landscape as it utilizes patients' own immune systems in combating the cancer cells. Cancer cells escape immune surveillance by hijacking the corresponding inhibitory pathways via overexpressing checkpoint genes. Phagocytosis checkpoints, such as CD47, CD24, MHC-I, PD-L1, STC-1 and GD2, have emerged as essential checkpoints for cancer immunotherapy by functioning as "don't eat me" signals or interacting with "eat me" signals to suppress immune responses. Phagocytosis checkpoints link innate immunity and adaptive immunity in cancer immunotherapy. Genetic ablation of these phagocytosis checkpoints, as well as blockade of their signaling pathways, robustly augments phagocytosis and reduces tumor size. Among all phagocytosis checkpoints, CD47 is the most thoroughly studied and has emerged as a rising star among targets for cancer treatment. CD47-targeting antibodies and inhibitors have been investigated in various preclinical and clinical trials. However, anemia and thrombocytopenia appear to be formidable challenges since CD47 is ubiquitously expressed on erythrocytes. Here, we review the reported phagocytosis checkpoints by discussing their mechanisms and functions in cancer immunotherapy, highlight clinical progress in targeting these checkpoints and discuss challenges and potential solutions to smooth the way for combination immunotherapeutic strategies that involve both innate and adaptive immune responses.
Collapse
|
8
|
Wu Y, Li Q, Lv LL, Chen JX, Ying HF, Ruan M, Zhu WH, Xu JY, Zhang CY, Zhang KY, Guo YB, Zhu WR, Zheng L. Nobiletin inhibits breast cancer cell migration and invasion by suppressing the IL-6-induced ERK-STAT and JNK-c-JUN pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154610. [PMID: 36584607 DOI: 10.1016/j.phymed.2022.154610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 11/16/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Breast cancer is one of the most common cancers in women, affecting more than 2 million women worldwide annually. However, effective treatments for breast cancer are limited. Nobiletin is a flavonoid present in the dried mature pericarp of mandarin orange (Citrus reticulata Blanco), which is used to prepare Citri Renetulatae Pericarpium and can inhibit tumour growth and progression according to modern pharmacological studies. However, whether nobiletin exhibits an antimetastatic role in breast cancer and its potential mechanism need to be further investigated. PURPOSE This study aims to evaluate the inhibitory effect of nobiletin on breast cancer and to elucidate potential mechanisms against invasion and migration. METHODS Cell viability was determined by cell counting kit-8 and colony formation assays. Wound healing and Boyden chamber assays detected cancer cell migration and invasion capabilities. Immunoblotting and qPCR were applied to determine the protein and mRNA expression levels of extracellular signal-regulated kinases (ERK) and the c-Jun N-terminal kinase (JNK) signalling pathways. Molecular docking was used to assess the degree of nobiletin binding to phosphatidylinositol 3-kinase (PI3K). Xenografts and liver metastases were constructed in BALB/c nude mice to evaluate the anticancer effect of nobiletin in vivo. H&E staining and immunohistochemistry were used to detect proliferation and the expression of related proteins. RESULTS Nobiletin induced cell death in a concentration- and time-dependent manner and possessed anti-invasion and anti-migration effects on MCF-7 and T47D cells by suppressing the interleukin-6-induced ERK and JNK signalling pathways. In addition, nobiletin docked with the binding site of PI3K, and the binding score was -8.0 kcal/mol. Furthermore, the inhibition of breast cancer growth and metastasis by nobiletin was demonstrated by constructing xenografts and liver metastases in vivo. CONCLUSION Nobiletin inhibited liver metastasis of breast cancer by downregulating the ERK-STAT and JNK-c-JUN pathways, and its safety and efficacy were verified, indicating the potential of nobiletin as an anticancer agent.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Qiong Li
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Ling-Ling Lv
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Jing-Xian Chen
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Hai-Feng Ying
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Ming Ruan
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Wen-Hua Zhu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Jia-Yue Xu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Chen-Yiyu Zhang
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Kai-Yuan Zhang
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Yuan-Biao Guo
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Wei-Rong Zhu
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Lan Zheng
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China.
| |
Collapse
|
9
|
Gao S, Zhang X, Liu J, Ji F, Zhang Z, Meng Q, Zhang Q, Han X, Wu H, Yin Y, Lv Y, Shi W. Icariin Induces Triple-Negative Breast Cancer Cell Apoptosis and Suppresses Invasion by Inhibiting the JNK/c-Jun Signaling Pathway. Drug Des Devel Ther 2023; 17:821-836. [PMID: 36969705 PMCID: PMC10038011 DOI: 10.2147/dddt.s398887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/04/2023] [Indexed: 03/29/2023] Open
Abstract
Background Breast cancer is a common cancer worldwide. Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer characterized by a poor prognosis. Icariin (ICA) is a flavonoid glycoside purified from the natural product Epimedium, which is reported to exert an inhibitory effect on a variety of cancers. However, molecular mechanisms behind ICA suppressed TNBC remain elusive. Methods The curative effects of ICA on TNBC cells and potential targets were predicted by network pharmacology and molecular biology methods screening, and the mechanism of inhibition was explained through in vitro experiments such as cell function determination, Western blot analysis, molecular docking verification, etc. Results This study showed that ICA inhibits TNBC cell functions such as proliferation, migration, and invasion in a dose-dependent manner. ICA could induce redox-induced apoptosis in TNBC cell, as shown by ROS upregulation. As a result of network pharmacology, ICA was predicted to be able to inhibit the MAPK signaling pathway. ICA treatment inhibited the expression of JNK and c-Jun and downregulated the antiapoptotic gene cIAP-2. Our results suggested that ICA could induce apoptosis by inducing an excessive accumulation of ROS in cells and suppress TNBC cell invasion via the JNK/c-Jun signaling pathway. Conclusion We demonstrated that ICA can effectively inhibit cell proliferation and induced apoptosis of TNBC cells. In addition, ICA could inhibit TNBC cell invasion through the JNK/c-Jun signaling pathway. The above suggests that ICA may become a potential drug for TNBC.
Collapse
Affiliation(s)
- Shenghan Gao
- The College of Life Sciences, Northwest University, Xi’an, 710069People’s Republic of China
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Xinyu Zhang
- The College of Life Sciences, Northwest University, Xi’an, 710069People’s Republic of China
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Jie Liu
- Clinical Medical Center, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Fuqing Ji
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Zhihao Zhang
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Qingjie Meng
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Qi Zhang
- The College of Life Sciences, Northwest University, Xi’an, 710069People’s Republic of China
| | - Xiaogang Han
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - He Wu
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Yulong Yin
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
| | - Yonggang Lv
- Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
- Correspondence: Yonggang Lv, Department of Thyroid Breast Surgery, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China, Email
| | - Wenzhen Shi
- Clinical Medical Center, Xi’an NO.3 Hospital, the Affiliated Hospital of Northwest University, Xi’an, People’s Republic of China
- Wenzhen Shi, Xi’an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi’an No.3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi’an, People’s Republic of China, Tel +8615037916770, Email
| |
Collapse
|
10
|
Huang S, Chen Y, Wu J, Chi Y. Development and validation of novel risk prediction models of breast cancer based on stanniocalcin‐1 level. Cancer Med 2022; 12:6499-6510. [PMID: 36336967 PMCID: PMC10067061 DOI: 10.1002/cam4.5419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/01/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The function of stanniocalcin-1 (STC-1) in the oncogenesis and progression of tumors has been extensively studied. The purpose of this study was to investigate the relationship between secreted STC-1 and prognosis in patients with breast cancer (BC) and to determine whether STC-1 could be a key prognostic factor in BC. METHODS The STC-1 level was measured by ELISA and clinical data from 1210 female patients with BC were used to develop and validate nomograms. We then verified the models through the plotting of ROC curves and calibration curves, calculating the C-index, and performing decision curve analyses (DCA). RESULTS The level of STC-1 in the peripheral plasma was significantly correlated with the T stage, N stage, clinical stage, grade, hormone receptors, HER-2 status, and tumor subtype. Cox regression analyses revealed that estrogen receptor(ER) status, N stage, and STC-1 level were risk factors for overall survival (OS), whereas T stage, N stage, and STC-1 level were independent prognostic factors for distant disease-free survival (DDFS) and disease-free survival (DFS). Both the ROC curve and the C-index confirmed the high resolution of these models, while the DCA identified the feasibility of their practical application. In addition, the calibration curves indicated good consistency between the predicted and actual survival rates. CONCLUSION Nomograms were created based on STC-1 levels for 3-, 5-, and 7-year OS, DDFS, and DFS of patients with BC respectively. As a key prognostic factor for BC, peripheral blood STC-1 level can be used clinically as a liquid biopsy indicator.
Collapse
Affiliation(s)
- Sheng Huang
- Department of Breast Surgery, Breast Cancer Institute Fudan University Shanghai Cancer Center, Fudan University Shanghai China
- The 2nd Department of Breast Surgery The Third Affiliated Hospital of Kunming Medical University Kunming China
| | - Yuyuan Chen
- The 2nd Department of Breast Surgery The Third Affiliated Hospital of Kunming Medical University Kunming China
- The Department of Thyroid and Breast Surgery The Affiliated Hospital of Ningbo University Medical College Ningbo China
| | - Jiong Wu
- Department of Breast Surgery, Breast Cancer Institute Fudan University Shanghai Cancer Center, Fudan University Shanghai China
| | - Yayun Chi
- Department of Breast Surgery, Breast Cancer Institute Fudan University Shanghai Cancer Center, Fudan University Shanghai China
| |
Collapse
|
11
|
Circular RNA circPOSTN promotes neovascularization by regulating miR-219a-2-3p/STC1 axis and stimulating the secretion of VEGFA in glioblastoma. Cell Death Dis 2022; 8:349. [PMID: 35927233 PMCID: PMC9352789 DOI: 10.1038/s41420-022-01136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/09/2022]
Abstract
Glioblastoma (GBM), the most malignant type of astrocytic tumor, is one of the deadliest cancers prevalent in adults. Along with tumor growth, patients with GBM generally suffer from extensive cerebral edema and apparent symptoms of intracranial hyper-pressure. Accumulating evidence has demonstrated that circRNA plays a critically important role in tumorigenesis and progression. However, the biological function and the underlying mechanism of circRNA in GBM remain elusive. In this study, by conducting gene expression detection based on 15 pairs of GBM clinical specimens and the normal adjunct tissues, we observed that circPOSTN showed abnormally higher expression in GBM. Both loss-of-function and gain-of-function biological experiments demonstrated that circPOSTN scheduled the proliferation, migration, and neovascularization abilities of GBM cells. Further, fluorescence in situ hybridization (FISH) assay, quantitative RT-PCR, and subcellular separation suggested that circPOSTN was predominately localized in the cytoplasm and may serve as a competing endogenous RNA (ceRNA). CircRNA-miRNA interaction prediction based on online analytical processing, AGO2-RIP assay, biotin labeled RNA pulldown assay, and dual-luciferase reporter assay revealed that circPOSTN sponged miR-219a-2-3p, limited its biological function, and ultimately upregulated their common downstream gene STC1. Finally, by carrying out in vitro and in vivo functional assays, we uncovered a new regulatory axis circPOSTN/miR-219a-2-3p/STC1 that promoted GBM neovascularization by increasing vascular endothelial growth factor A (VEGFA) secretion. Our study underscores the critical role of circPOSTN in GBM progression, providing a novel insight into GBM anti-tumor therapy.
Collapse
|
12
|
Comprehensive Analyses of Stromal-Immune Score-Related Competing Endogenous RNA Networks In Colon Adenocarcinoma. DISEASE MARKERS 2022; 2022:4235305. [PMID: 35607443 PMCID: PMC9124109 DOI: 10.1155/2022/4235305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/12/2022] [Accepted: 04/21/2022] [Indexed: 12/14/2022]
Abstract
Although recent clinical investigations emphasize the roles of myriad diversities of RNAs in stromal and immune components in the tumor microenvironment, especially in colon adenocarcinoma, however, analyses of “competing endogenous RNAs (ceRNA)” network in association with stromal and immune scores have yet to be determined. This study was conducted to explore the regulatory mechanisms of a stromal-immune score-based ceRNA network in colon adenocarcinoma. Stromal and immune scores of colon adenocarcinoma tumor samples were calculated by using the ESTIMATE algorithm. Differential expression analysis between samples with high/low stromal and immune scores was performed, followed by functional annotation for the overlapping DEmRNAs. The ceRNA network was constructed by differential expression analysis, prediction of RNA-RNA interaction, and correlation with clinicopathological parameters of the patients, which were further verified by external datasets and experiments. Colon adenocarcinoma patients having higher immune scores exhibited prolonged overall survival. RNA dataset analyses from TCGA revealed aberrant expressions of a total of 2052 mRNAs, 108 lncRNAs, and 70 miRNAs between high and low stromal/immune groups. Functional annotation mapped the differentially overexpressed mRNAs for immune-associated GO terms. To construct the ceRNA network, a total of 48 lncRNAs, 40 miRNAs, and 199 mRNAs were sorted out. A dysregulated ceRNA network consisting of 6 lncRNAs, 11 miRNAs, and 39 mRNAs was constructed by comparing RNA expressions between cancer as well as adjacent normal tissues. The ceRNA regulatory axis “MIAT/miR-532-3p/STC1” was regarded as a potential hit by the comprehensive analysis. The RT-qPCR assay showed upregulation of MIAT and STC1 while downregulation of hsa-miR-532-3p expression in cancer. Thus, our study highlights the potential role of a stromal-immune score-based ceRNA network in the colon adenocarcinoma microenvironment. The ceRNA axis MIAT/miR-532-3p/STC1 could serve as a promising therapeutic target for colon adenocarcinoma.
Collapse
|
13
|
Serum Autoantibodies against LRDD, STC1, and FOXA1 as Biomarkers in the Detection of Ovarian Cancer. DISEASE MARKERS 2022; 2022:6657820. [PMID: 35273656 PMCID: PMC8904091 DOI: 10.1155/2022/6657820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 10/03/2021] [Accepted: 01/29/2022] [Indexed: 01/13/2023]
Abstract
Purpose This study is aimed at evaluating serum autoantibodies against four tumor-associated antigens, including LRDD, STC1, FOXA1, and EDNRB, as biomarkers in the immunodiagnosis of ovarian cancer (OC). Methods The autoantibodies against LRDD, STC1, FOXA1, and EDNRB were measured using an enzyme-linked immunosorbent assay (ELISA) in 94 OC patients and 94 normal healthy controls (NHC) in the research group. In addition, the diagnostic values of different autoantibodies were validated in another independent validation group, which comprised 136 OC patients, 136 NHC, and 181 patients with benign ovarian diseases (BOD). Results In the research group, autoantibodies against LRDD, STC1, and FOXA1 had higher serum titer in OC patients than NHC (P < 0.001). The area under receiver operating characteristic curves (AUCs) of these three autoantibodies were 0.910, 0.879, and 0.817, respectively. In the validation group, they showed AUCs of 0.759, 0.762, and 0.817 and sensitivities of 49.3%, 42.7%, and 48.5%, respectively, at specificity over 90% for discriminating OC patients from NHC. For discriminating OC patients from BOD, they showed AUCs of 0.718, 0.729, and 0.814 and sensitivities of 47.1%, 39.0%, and 51.5%, respectively, at specificity over 90%. The parallel analyses demonstrated that the combination of anti-LRDD and anti-FOXA1 autoantibodies achieved the optimal diagnostic performance with the sensitivity of 58.1% at 87.5% specificity and accuracy of 72.8%. The positive rate of the optimal autoantibody panel improved from 62.4% to 87.1% when combined with CA125 in detecting OC patients. Conclusion Serum autoantibodies against LRDD, STC1, and FOXA1 have potential diagnostic values in detecting OC.
Collapse
|
14
|
Hou J, Cheng J, Dai Z, Wei N, Chen H, Wang S, Dai M, Li L, Wang H, Ni Q. Molecular and Clinical Significance of Stanniocalcin-1 Expression in Breast Cancer Through Promotion of Homologous Recombination-Mediated DNA Damage Repair. Front Cell Dev Biol 2021; 9:731086. [PMID: 34722511 PMCID: PMC8554131 DOI: 10.3389/fcell.2021.731086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/20/2021] [Indexed: 11/23/2022] Open
Abstract
Stanniocalcin-1 (STC1) is a glycoprotein hormone whose abnormal expression has been reported to be associated with a variety of tumors, but its function in breast cancer is not well understood. Through modulation of STC1 expression in different breast cancer cell lines, our study found that STC1 could promote the proliferation and growth of breast cancer cells and promote metastasis. Furthermore, STC1 reduced apoptosis induction by irradiation. We also found that STC1 could promote a homologous recombination-mediated DNA damage repair by recruiting BRCA1 to sites of damage. Moreover, STC1 silencing sensitized breast cancer cells to treatment with irradiation (IR), olaparib, or cisplatin in vitro. In clinical settings, the serum concentration of STC1 was higher in breast cancer patients than in healthy women, as detected by enzyme-linked immunosorbent assay (ELISA). In addition, immunohistochemical staining of breast cancer specimens showed that a high expression of STC1 was negatively correlated with recurrence-free survival in breast cancer, indicating that STC1 expression could be used as a predictive marker for a poor prognosis in breast cancer. All these findings indicate that STC1 promotes breast cancer tumorigenesis and that breast cancers with a high level of STC1 are more resistant to treatment, probably through homologous recombination (HR) promotion. Furthermore, combining STC1 inhibition and DNA damage-inducing drugs may be a novel approach to improve the survival of patients with STC1-expressing breast cancer.
Collapse
Affiliation(s)
- Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jigan Cheng
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - ZeHua Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Na Wei
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Huan Chen
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shu Wang
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Min Dai
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Leilei Li
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hua Wang
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qing Ni
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
15
|
Hu S, Liu H, Zhang J, Li S, Zhou H, Gao Y. Effects and prognostic values of miR-30c-5p target genes in gastric cancer via a comprehensive analysis using bioinformatics. Sci Rep 2021; 11:20584. [PMID: 34663825 PMCID: PMC8523699 DOI: 10.1038/s41598-021-00043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/30/2021] [Indexed: 12/02/2022] Open
Abstract
Gastric cancer (GC) is a common cancer and the leading cause of cancer-related death worldwide. To improve the diagnosis and treatment of GC, it is necessary to identify new biomarkers by investigating the cellular and molecular mechanisms. In this study, miR-30c-5p expression was significantly down-regulated in GC tissues by comprehensive analysis using multiple databases. The target genes of miR-30c-5p with up-regulated expression level in GC were identified, including ADAM12 (a disintegrin and metalloproteinase12), EDNRA (the Endothelin receptor type A), STC1 (stanniocalcin 1), and CPNE8 (the calcium-dependent protein, copine 8). The expression level of ADAM12 was significantly related to depth of invasion (p = 0.036) in GC patients. The expression level of EDNRA was significantly related to grade (P = 0.003), depth of invasion (P = 0.019), and lymphatic metastasis (P = 0.001). The expression level of CPNE8 was significantly related to grade (P = 0.043) and TNM stage (P = 0.027).Gene set enrichment analysis showed that they might participate in GC progression through cancer-related pathways. CIBERSORT algorithm analysis showed that their expressions were related to a variety of tumor-infiltrating immune cells. The higher expression of those target genes might be the independent risk factor for poor survival of GC patients, and they might be potential prognostic markers in GC patients.
Collapse
Affiliation(s)
- Shangshang Hu
- Research Center of Clinical Laboratory Science, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Huaifeng Liu
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Jinyan Zhang
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China.,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Shujing Li
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Huadong Zhou
- Department of Neurology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.,Department of Neurology, Army Medical Center of PLA, Chongqing, 400038, China
| | - Yu Gao
- School of Life Science, Bengbu Medical College, Bengbu, 233030, Anhui, China. .,Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, 233030, Anhui, China. .,School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, No. 2600 Donghai Road, Bengbu, 233030, Anhui, China.
| |
Collapse
|
16
|
Khatun M, Urpilainen E, Ahtikoski A, Arffman RK, Pasanen A, Puistola U, Tapanainen JS, Andersson LC, Butzow R, Loukovaara M, Piltonen TT. Low Expression of Stanniocalcin 1 (STC-1) Protein Is Associated With Poor Clinicopathologic Features of Endometrial Cancer. Pathol Oncol Res 2021; 27:1609936. [PMID: 34650342 PMCID: PMC8505533 DOI: 10.3389/pore.2021.1609936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022]
Abstract
Stanniocalcin-1 (STC-1) is a glycoprotein hormone involved in diverse biological processes, including regulation of calcium phosphate homeostasis, cell proliferation, apoptosis, inflammation, oxidative stress responses, and cancer development. The role of STC-1 in endometrial cancer (EC) is yet to be elucidated. In this study, we investigated the protein expression pattern of STC-1 in a tissue microarray (TMA) cohort of hysterectomy specimens from 832 patients with EC. We then evaluated the prognostic value of STC-1 expression regarding the clinicopathologic features and patients survival over a period of 140 months. Our results revealed that in EC tissue samples, STC-1 is mainly localized in the endometrial epithelium, although some expression was also observed in the stroma. Decreased STC-1 expression was associated with factors relating to a worse prognosis, such as grade 3 endometrioid tumors (p = 0.030), deep myometrial invasion (p = 0.003), lymphovascular space invasion (p = 0.050), and large tumor size (p = 0.001). Moreover, STC-1 expression was decreased in tumors obtained from obese women (p = 0.014) and in women with diabetes mellitus type 2 (DMT2; p = 0.001). Interestingly, the data also showed an association between DNA mismatch repair (MMR) deficiency and weak STC-1 expression, specifically in the endometrial epithelium (p = 0.048). No association was observed between STC-1 expression and disease-specific survival. As STC-1 expression was particularly low in cases with obesity and DMT2 in the TMA cohort, we also evaluated the correlation between metformin use and STC-1 expression in an additional EC cohort that only included women with DMT2 (n = 111). The analysis showed no difference in STC-1 expression in either the epithelium or the stroma in women undergoing metformin therapy compared to metformin non-users. Overall, our data may suggest a favorable role for STC-1 in EC behavior; however, further studies are required to elucidate the detailed mechanism and possible applications to cancer treatment.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Elina Urpilainen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Anne Ahtikoski
- Department of Pathology, Oulu University Hospital, University of Oulu, Oulu, Finland.,Department of Pathology, Turku University Hospital, Turku, Finland
| | - Riikka K Arffman
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Annukka Pasanen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Ulla Puistola
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland.,Department of Obstetrics and Gynaecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Leif C Andersson
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Ralf Butzow
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mikko Loukovaara
- Department of Obstetrics and Gynaecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Terhi T Piltonen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
17
|
Sun J, Wei X, You J, Yue W, Ouyang J, Ling Z, Hou J. STC1 is a Novel Biomarker Associated with Immune Characteristics and Prognosis of Bladder Cancer. Int J Gen Med 2021; 14:5505-5516. [PMID: 34539184 PMCID: PMC8445105 DOI: 10.2147/ijgm.s329723] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Background Stanniocalcin-1 (STC1) is a well-studied oncogene that promotes different types of cancer progression. However, the expression status of STC1, the values of STC1 on prognosis, and its immune characteristic in bladder cancer (BLCA) have not been well examined. Methods The expression of STC1 and its clinicopathological as well as immune characteristics in BLCA samples were firstly identified in The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. Immunohistochemistry (IHC) performed on the tissue microarray (TMA) slide was further used to validate the expression of STC1 and its relationship with immune features in 16 non-muscle invasive bladder cancer (NMIBC) samples and 42 muscle invasive bladder cancer (MIBC) samples. Results The expression of STC1 was upregulated in higher stage BLCA. High STC1 expression also predicted poor prognosis in BLCA. Subsequently, the TMA validated the expression and prognostic value of STC1 in BLCA. Bioinformatics analysis demonstrated that STC1 and common immune checkpoints as well as immune markers of various immune cells were positively correlated in TCGA. In addition, IHC data from the TMA further validated that tumor cells with higher STC1 level tended to express higher PDL1 as well as increased infiltration of CD3+ T cells. Conclusion To our knowledge, this is the first comprehensive study that investigates the clinical and immune characteristics of STC1 in BLCA. It may provide new insight into the function of STC1 in regulating tumor immune microenvironment. Further studies are warranted to uncover the potential mechanisms that mediate STC1 expression and tumor immunity in BLCA.
Collapse
Affiliation(s)
- Jiale Sun
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xuedong Wei
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jiawei You
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Wenchang Yue
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Ouyang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhixin Ling
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
18
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
19
|
Xu X, Nie J, Lu L, Du C, Meng F, Song D. YAP-TEAD up-regulates IRS2 expression to induce and deteriorate oesophageal cancer. J Cell Mol Med 2021; 25:2584-2595. [PMID: 33570213 PMCID: PMC7933937 DOI: 10.1111/jcmm.16266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 12/07/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Oesophageal cancer (EC) represents a significant cause of cancer worldwide. Yes‐associated protein (YAP) is reported to correlate with the initiation of multiple cancers including EC, but the underlying mechanism remains elusive. The current study aimed to investigate the molecular mechanism of YAP‐TEAD in the occurrence and progression of EC. EC tissues and cells were obtained, followed by determination of the expression of YAP, c‐Jun, pc‐Jun and IRS2. The effect of YAP‐TEAD on the biological EC cell processes was explored through gain‐ and loss‐of‐function approaches. The interaction between YAP and TEAD was detected by co‐immunoprecipitation. The binding of TEAD to the c‐Jun promoter was determined using chromatin immunoprecipitation. Tumour formation in the nude mice was detected in order to ascertain the effect of YAP and IRS2 in vivo. We found elevated YAP in the EC tissues and cells. YAP silencing led to a decrease in EC cell proliferation, invasion and sphere formation. YAP‐TEAD complex bound to the promotor of c‐Jun, and c‐Jun led to an increase in the expression of IRS2 through the JNK/c‐Jun pathway. Additionally, pc‐Jun and phosphorylated JNK were localized in the nuclear in addition to displaying enhanced expression in the EC tissues. IRS2 overexpression negated the inhibition of cell proliferation, invasion and sphere formation triggering YAP silencing. YAP up‐regulated IRS2 and aggravated EC in vivo. Taken together, YAP‐TEAD activates the JNK/c‐Jun pathway to up‐regulate IRS2, ultimately promoting EC progression. Therefore, YAP‐TEAD inhibition could be a promising therapeutic approach for EC treatment.
Collapse
Affiliation(s)
- Xiangming Xu
- Department of Gastroenterology, Linyi People's Hospital, Linyi, China
| | - Jiao Nie
- Department of Gastroenterology, Linyi People's Hospital, Linyi, China
| | - Lin Lu
- Department of Gastroenterology, Linyi People's Hospital, Linyi, China
| | - Chao Du
- Department of Gastroenterology, Linyi People's Hospital, Linyi, China
| | - Fansheng Meng
- Department of Gastroenterology, Linyi People's Hospital, Linyi, China
| | - Duannuo Song
- Department of Gastroenterology, Linyi People's Hospital, Linyi, China
| |
Collapse
|
20
|
Yamagishi T, Kawashima H, Ogose A, Ariizumi T, Oike N, Sasaki T, Hatano H, Endo N. <Editors' Choice> Stanniocalcin-1 mRNA expression in soft-tissue tumors. NAGOYA JOURNAL OF MEDICAL SCIENCE 2021; 82:85-92. [PMID: 32273636 PMCID: PMC7103862 DOI: 10.18999/nagjms.82.1.85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stanniocalcin-1 (STC1) is a glycoprotein that was originally identified as a calcium-regulating hormone in bony fish, and that has been shown to also critically mediate cell growth, proliferation and differentiation, etc. in humans. Increased STC1 expression levels have been previously detected in different human cancer samples, such as those isolated from lung, breast, ovary, colon, pancreas, and liver tumors; thus, the present study evaluated STC1 expression in various soft-tissue tumors. STC1 mRNA isolated from 16 cell lines and 186 clinical soft-tissue tumor specimens were analyzed via quantitative real-time PCR, and the calculated expression levels were normalized to those exhibited by STC1-expressing MDA-MB-231 cells. The results of these analyses did not reveal any specific histological tumor types that displayed significantly increased STC1 expression; however, they did not indicate that STC1 expression was significantly higher in malignant compared to benign soft-tissue tumors. Furthermore, in adipocytic tumors, STC1 expression in dedifferentiated liposarcomas was found to be highest and lowest in lipoma tissues, respectively, suggesting that adipocytic tumors may express increasely high levels of STC1 mRNA as they become histologically more advanced. STC1 expression correlates with the malignancy grade in soft-tissue tumors.
Collapse
Affiliation(s)
- Tetsuro Yamagishi
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Kawashima
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Akira Ogose
- Department of Orthopedic Surgery, Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, Minami-Uonuma, Japan
| | - Takashi Ariizumi
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Oike
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Taro Sasaki
- Department of Orthopedic Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hiroshi Hatano
- Department of Orthopedic Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Naoto Endo
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
21
|
Pereira M, Matuszewska K, Jamieson C, Petrik J. Characterizing Endocrine Status, Tumor Hypoxia and Immunogenicity for Therapy Success in Epithelial Ovarian Cancer. Front Endocrinol (Lausanne) 2021; 12:772349. [PMID: 34867818 PMCID: PMC8635771 DOI: 10.3389/fendo.2021.772349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer is predominantly diagnosed at advanced stages which creates significant therapeutic challenges. As a result, the 5-year survival rate is low. Within ovarian cancer, significant tumor heterogeneity exists, and the tumor microenvironment is diverse. Tumor heterogeneity leads to diversity in therapy response within the tumor, which can lead to resistance or recurrence. Advancements in therapy development and tumor profiling have initiated a shift from a "one-size-fits-all" approach towards precision patient-based therapies. Here, we review aspects of ovarian tumor heterogeneity that facilitate tumorigenesis and contribute to treatment failure. These tumor characteristics should be considered when designing novel therapies or characterizing mechanisms of treatment resistance. Individual patients vary considerably in terms of age, fertility and contraceptive use which innately affects the endocrine milieu in the ovary. Similarly, individual tumors differ significantly in their immune profile, which can impact the efficacy of immunotherapies. Tumor size, presence of malignant ascites and vascular density further alters the tumor microenvironment, creating areas of significant hypoxia that is notorious for increasing tumorigenesis, resistance to standard of care therapies and promoting stemness and metastases. We further expand on strategies aimed at improving oxygenation status in tumors to dampen downstream effects of hypoxia and set the stage for better response to therapy.
Collapse
|
22
|
Liu JH, Cao YM, Rong ZP, Ding J, Pan X. Trichostatin A Induces Autophagy in Cervical Cancer Cells by Regulating the PRMT5-STC1-TRPV6-JNK Pathway. Pharmacology 2020; 106:60-69. [PMID: 33142290 DOI: 10.1159/000507937] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/15/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the effects of trichostatin A (TSA) on cervical cancer and the related mechanisms. METHODS The HeLa and Caski cervical cancer cell lines were treated with different concentrations of TSA. Cell viability was measured by MTT assays. Cell apoptosis was analysed using flow cytometry. Expression of transient receptor potential cation channel, subfamily V, member 6 (TRPV6), protein arginine methyltransferase 5 (PRMT5), and stanniocalcin 1 (STC1) was determined by qRT-PCR and Western blotting. Protein levels of LC3 II/I, beclin1, p62, JNK, and p-JNK were detected by Western blotting. RESULTS Treatment with TSA significantly decreased HeLa and Caski cell viability and enhanced the apoptosis rate in a dose-dependent manner. TSA markedly elevated beclin1 protein levels and the LC3 II/I ratio and significantly reduced p62 levels in a dose-dependent manner. In addition, TSA (1 μM) significantly suppressed PRMT5 and TRPV6 levels and enhanced STC1 and p-JNK levels. The lysosomal inhibitor bafilomycin-A1 synergistically enhanced the TSA-mediated increase in autophagic flux. Either the overexpression of TRPV6 or the inhibition of JNK signalling markedly enhanced cell viability, inhibited apoptosis, and autophagy and reduced p-JNK levels in TSA-treated cells. The inhibition of STC1 significantly increased TRPV6 protein levels and reduced p-JNK levels. Overexpression of PRMT5 dramatically decreased STC1 and p-JNK protein levels and increased TRPV6 levels. CONCLUSION TSA suppresses cervical cancer cell proliferation and induces apoptosis and autophagy through regulation of the PRMT5/STC1/TRPV6/JNK axis.
Collapse
Affiliation(s)
- Jian-Hao Liu
- School of Pharmaceutical Sciences of Central South University, Changsha, China
| | - Yan-Ming Cao
- Department of Oncology, Xiangya Third Hospital, Central South University, Changsha, China
| | - Zhi-Peng Rong
- Department of Oncology, Xiangya Third Hospital, Central South University, Changsha, China
| | - Juan Ding
- Department of Oncology, Xiangya Third Hospital, Central South University, Changsha, China
| | - Xi Pan
- Department of Oncology, Xiangya Third Hospital, Central South University, Changsha, China,
| |
Collapse
|
23
|
Yang L, Zhu X, Ni Y, Wu D, Tian Y, Chen Z, Li M, Zhang H, Liang D. MicroRNA-34c Inhibits Osteogenic Differentiation and Valvular Interstitial Cell Calcification via STC1-Mediated JNK Pathway in Calcific Aortic Valve Disease. Front Physiol 2020; 11:829. [PMID: 32982764 PMCID: PMC7489094 DOI: 10.3389/fphys.2020.00829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 06/22/2020] [Indexed: 12/20/2022] Open
Abstract
Calcific aortic valve disease (CAVD), a common heart valve disease, is increasingly prevalent worldwide and causes high morbidity and mortality. Here, we aimed to investigate a possible role for miR-34c in the development of osteogenic differentiation during CAVD and to find out the underlying mechanisms. Valvular interstitial cells (VICs) were isolated from the clinical aortic valve tissue samples of CAVD patients and patients with acute aortic dissection and collected. Then, RT-qPCR was performed to determine miR-34c expression and western blot analysis was applied to confirm the relevant protein expression in these VICs. Dual luciferase reporter gene assay was applied to confirm the relation between miR-34c and STC1. Alkaline phosphatase (ALP) staining and alizarin red staining was performed to further confirm the degree of calcification in these samples. MiR-34c was lowly expressed and STC1 was highly expressed in the CAVD tissues. Furthermore, STC1 was the target of miR-34c and was negatively regulated by miR-34c. Overexpression of miR-34c in VICs was concomitant with suppression of both STC1 expression and phosphorylation level of c-Jun N-terminal kinase (JNK). In addition, significant decrease of bone morphogenetic protein-2 (BMP2) and osteocalcin, as well as the decrease of calcification degree were also observed in VICs with miR-34c overexpressed. Taken together, miR-34c could inhibit osteogenic differentiation and calcification of VICs by suppressing the STC1/JNK signaling pathway in CAVD, making miR-34c a novel therapeutic target for the treatment of CAVD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Degang Liang
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
24
|
Zhao F, Yang G, Feng M, Cao Z, Liu Y, Qiu J, You L, Zheng L, Zhang T, Zhao Y. Expression, function and clinical application of stanniocalcin-1 in cancer. J Cell Mol Med 2020; 24:7686-7696. [PMID: 32468698 PMCID: PMC7348177 DOI: 10.1111/jcmm.15348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/10/2019] [Accepted: 10/19/2019] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein stanniocalcin-1 functions as a regulatory endocrine hormone that maintains the balance of calcium and phosphorus in bony fish and as a paracrine/autocrine factor involved in many physiological/pathological processes in humans, including carcinogenesis. In this review, we provide an overview of (a) the possible mechanisms through which STC1 affects the malignant properties of cancer, (b) transcriptional and post-transcriptional regulation pathways of STC1 and (c) the potential clinical relevance of STC1 as a cancer biomarker and even a therapeutic target in the future. Exploring the role of STC1 in cancer development may provide a better understanding of the tumorigenesis process in humans and may facilitate finding an effective therapeutic method against cancer.
Collapse
Affiliation(s)
- Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Cernea A, Fernández-Martínez JL, deAndrés-Galiana EJ, Fernández-Ovies FJ, Alvarez-Machancoses O, Fernández-Muñiz Z, Saligan LN, Sonis ST. Robust pathway sampling in phenotype prediction. Application to triple negative breast cancer. BMC Bioinformatics 2020; 21:89. [PMID: 32164540 PMCID: PMC7068866 DOI: 10.1186/s12859-020-3356-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background Phenotype prediction problems are usually considered ill-posed, as the amount of samples is very limited with respect to the scrutinized genetic probes. This fact complicates the sampling of the defective genetic pathways due to the high number of possible discriminatory genetic networks involved. In this research, we outline three novel sampling algorithms utilized to identify, classify and characterize the defective pathways in phenotype prediction problems, such as the Fisher’s ratio sampler, the Holdout sampler and the Random sampler, and apply each one to the analysis of genetic pathways involved in tumor behavior and outcomes of triple negative breast cancers (TNBC). Altered biological pathways are identified using the most frequently sampled genes and are compared to those obtained via Bayesian Networks (BNs). Results Random, Fisher’s ratio and Holdout samplers were more accurate and robust than BNs, while providing comparable insights about disease genomics. Conclusions The three samplers tested are good alternatives to Bayesian Networks since they are less computationally demanding algorithms. Importantly, this analysis confirms the concept of “biological invariance” since the altered pathways should be independent of the sampling methodology and the classifier used for their inference. Nevertheless, still some modifications are needed in the Bayesian networks to be able to sample correctly the uncertainty space in phenotype prediction problems, since the probabilistic parameterization of the uncertainty space is not unique and the use of the optimum network might falsify the pathways analysis.
Collapse
Affiliation(s)
- Ana Cernea
- Group of Inverse Problems, Optimization and Machine Learning, Department of Mathematics, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain
| | - Juan Luis Fernández-Martínez
- Group of Inverse Problems, Optimization and Machine Learning, Department of Mathematics, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain.
| | - Enrique J deAndrés-Galiana
- Group of Inverse Problems, Optimization and Machine Learning, Department of Mathematics, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain.,Department of Informatics and Computer Science, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain
| | - Francisco Javier Fernández-Ovies
- Group of Inverse Problems, Optimization and Machine Learning, Department of Mathematics, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain
| | - Oscar Alvarez-Machancoses
- Group of Inverse Problems, Optimization and Machine Learning, Department of Mathematics, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain
| | - Zulima Fernández-Muñiz
- Group of Inverse Problems, Optimization and Machine Learning, Department of Mathematics, University of Oviedo, C/ Federico García-Lorca, 18, 33007, Oviedo, Spain
| | - Leorey N Saligan
- National Institutes of Health, National Institute of Nursing Research, Bethesda, MD, USA
| | - Stephen T Sonis
- Primary Endpoint Solutions, Watertown, MA, USA.,Brigham and Women's Hospital and the Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
26
|
Al-Abdallah A, Jahanbani I, Mehdawi H, Ali RH, Al-Brahim N, Mojiminiyi O. The stress-activated protein kinase pathway and the expression of stanniocalcin-1 are regulated by miR-146b-5p in papillary thyroid carcinogenesis. Cancer Biol Ther 2020; 21:412-423. [PMID: 32037949 PMCID: PMC7515490 DOI: 10.1080/15384047.2020.1721250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Papillary thyroid cancer (PTC) is the most common type of thyroid cancer. Deciphering the pathophysiological mechanisms that contribute to PTC development is essential to the discovery of optimal diagnostic and therapeutic approaches. MiR-146b-5p has been identified as a cancer-associated microRNA highly up-regulated in PTC. This study explores the hypothesis that miR-146b-5p contributes to papillary thyroid carcinogenesis through regulation of cell signaling pathways in a manner that overcomes the cellular growth suppressive events and provides survival advantage. The effect of miR-146b-5p inhibition on major cancer related signaling pathways and expression of Stanniocalcin-1 (STC1), an emerging molecule associated with stress response and carcinogenesis, was tested in cultured primary thyroid cells using luciferase reporter assays, quantitative real-time PCR, immunofluorescence staining, and flow cytometry. Our results demonstrated that miR-146b-5p inhibits the JNK/AP1 pathway activity and down-regulates the expression of STC-1 in thyroid-cultured cells and in thyroid tissue samples. In the presence of miR-146b-5p, PTC cells were resistant to cell death in response to oxidative stress. This is a novel report that miR-146b-5p directly targets STC1 and regulates the activity of JNK/AP1 pathway. Considering the importance of the JNK/AP1 pathway and STC1 in mediating many physiological and pathological processes like apoptosis, stress response and cellular metabolism, a biological regulator of these pathways would have a great scientific and clinical significance.
Collapse
Affiliation(s)
| | - Iman Jahanbani
- Pathology Department, Kuwait University, Kuwait City, Kuwait
| | - Heba Mehdawi
- Pathology Department, Kuwait University, Kuwait City, Kuwait
| | - Rola H Ali
- Pathology Department, Kuwait University, Kuwait City, Kuwait
| | | | | |
Collapse
|
27
|
Wu Q, Wu W, Jacevic V, Franca TCC, Wang X, Kuca K. Selective inhibitors for JNK signalling: a potential targeted therapy in cancer. J Enzyme Inhib Med Chem 2020; 35:574-583. [PMID: 31994958 PMCID: PMC7034130 DOI: 10.1080/14756366.2020.1720013] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
c-Jun N-terminal kinase (JNK) signalling regulates both cancer cell apoptosis and survival. Emerging evidence show that JNK promoted tumour progression is involved in various cancers, that include human pancreatic-, lung-, and breast cancer. The pro-survival JNK oncoprotein functions in a cell context- and cell type-specific manner to affect signal pathways that modulate tumour initiation, proliferation, and migration. JNK is therefore considered a potential oncogenic target for cancer therapy. Currently, designing effective and specific JNK inhibitors is an active area in the cancer treatment. Some ATP-competitive inhibitors of JNK, such as SP600125 and AS601245, are widely used in vitro; however, this type of inhibitor lacks specificity as they indiscriminately inhibit phosphorylation of all JNK substrates. Moreover, JNK has at least three isoforms with different functions in cancer development and identifying specific selective inhibitors is crucial for the development of targeted therapy in cancer. Some selective inhibitors of JNK are identified; however, their clinical studies in cancer are relatively less conducted. In this review, we first summarised the function of JNK signalling in cancer progression; there is a focus on the discussion of the novel selective JNK inhibitors as potential targeting therapy in cancer. Finally, we have offered a future perspective of the selective JNK inhibitors in the context of cancer therapies. We hope this review will help to further understand the role of JNK in cancer progression and provide insight into the design of novel selective JNK inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China.,College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Wenda Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Vesna Jacevic
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,National Poison Control Centre, Military Medical Academy, Belgrade, Serbia.,Medical Faculty of the Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Tanos C C Franca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Laboratory of Molecular Modeling Applied to the Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro, Brazil
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, China
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
28
|
Chen F, Zhang Z, Pu F. Role of stanniocalcin-1 in breast cancer. Oncol Lett 2019; 18:3946-3953. [PMID: 31579413 PMCID: PMC6757304 DOI: 10.3892/ol.2019.10777] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 07/16/2019] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is a highly heterogeneous disease consisting of five disease subtypes with distinct histological characteristics, clinical behaviors and prognostic features. Stanniocalcin-1 (STC1) is a secreted glycoprotein hormone that has been demonstrated to regulate calcium and phosphate homeostasis. Mammalian STC1 is expressed in various tissues and is implicated in multiple physiological and pathophysiological processes. In addition, growing evidence has suggested that STC1 serves an oncogenic role in a number of different types of tumor. However, the role of STC1 in breast cancer is complex, considering that some studies have shown that it exerts an oncogenic role, whereas other studies have demonstrated the opposite. The aim of the present review article is to evaluate the currently available data on mammalian STC1 and discuss its potential roles in each subtype of breast cancer.
Collapse
Affiliation(s)
- Fengxia Chen
- Department of Medical Oncology, General Hospital of The Yangtze River Shipping, Wuhan, Hubei 430010, P.R. China.,Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Feifei Pu
- Department of Orthopedics, Wuhan No. 1 Hospital, Wuhan Integrated Traditional Chinese Medicine and Western Medicine Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
29
|
Xiong Y, Wang Q. STC1 regulates glioblastoma migration and invasion via the TGF‑β/SMAD4 signaling pathway. Mol Med Rep 2019; 20:3055-3064. [PMID: 31432189 PMCID: PMC6755173 DOI: 10.3892/mmr.2019.10579] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/01/2019] [Indexed: 02/06/2023] Open
Abstract
Stanniocalcin-1 (STC1) is involved in cancer progression; however, the function of STC1 in glioblastoma remains unknown. In the present study, the expression levels of STC1 protein in glioblastoma were detected using immunohistochemistry. The expression levels of STC1, SMAD2/3 and SMAD4 proteins, following silencing of STC1, were assessed via western blotting. EdU and Transwell assays were performed to determine the proliferation and migration ability of the cells. The mRNA expression levels of STC1, SMAD4 and microRNA (miR)-34a were determined using quantitative PCR. The expression levels of STC1 were increased in glioblastoma tissues. STC1 revealed a significant association with poor outcome in patients with glioblastoma (P<0.05). The proliferation and invasion abilities were repressed in LN229 cells infected with LV3-shSTC1-1 and LV3-shSTC1-2 compared with LV3-NC. By contrast, the proliferation and invasion abilities were increased in T98G cells infected with LV5-STC1 compared with LV5-NC (P<0.05). The expression levels of STC1, SMAD2/3 and SMAD4 were decreased in LN229 cells infected with LV3-shSTC1-1 and LV3-shSTC1-2 compared with LV3-NC. However, the expression levels of STC1, SMAD2/3 and SMAD4 were elevated in T98G cells infected with LV5-STC1 compared with LV5-NC. The expression levels of miR-34a were decreased following silencing of STC1 (P<0.05). The expression levels of SMAD4 were decreased when transfected with miR-34a mimics (P<0.05). The luciferase activity of the wild-type 3′untranslated region of SMAD4 was decreased following transfection with miR-34a mimics (P<0.05). Silencing of STC1 inhibited the growth of LN229 in vivo. In conclusion, STC1 expression levels were increased in the present study, and it was revealed that STC1 regulated glioblastoma malignancy. This phenotype was observed in the SMAD2/3 and SMAD4 pathways.
Collapse
Affiliation(s)
- Yan Xiong
- Department of Neurosurgery, Chongqing Ninth People's Hospital, Chongqing 400715, P.R. China
| | - Qibai Wang
- Department of Neurosurgery, Chongqing Red Cross Hospital (People's Hospital of Jiangbei District), Chongqing 400020, P.R. China
| |
Collapse
|
30
|
Mao Q, Nguyen PD, Shanti RM, Shi S, Shakoori P, Zhang Q, Le AD. Gingiva-Derived Mesenchymal Stem Cell-Extracellular Vesicles Activate Schwann Cell Repair Phenotype and Promote Nerve Regeneration. Tissue Eng Part A 2019; 25:887-900. [DOI: 10.1089/ten.tea.2018.0176] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Qin Mao
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P.R. China
| | - Phuong D. Nguyen
- Division of Plastic and Reconstructive Surgery, University of Pennsylvania Perelman School of Medicine and Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rabie M. Shanti
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
- Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Otolaryngology and Head and Neck Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shihong Shi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Pasha Shakoori
- Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
| | - Anh D. Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania
- Department of Oral and Maxillofacial Surgery, Perelman Center for Advanced Medicine, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Zhu Y, Pan Y, Zhang G, Wu Y, Zhong W, Chu C, Qian Y, Zhu G. Chelerythrine Inhibits Human Hepatocellular Carcinoma Metastasis in Vitro. Biol Pharm Bull 2017; 41:36-46. [PMID: 29093327 DOI: 10.1248/bpb.b17-00451] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chelerythrine (CHE) is a type of benzophenanthridine alkaloid found in many herbs and is also the main alkaloid constituent of Toddalia asiatica (L.) LAM. It has been proven to have various activities including antitumor, antifungal, anti-inflammatory and anti-parasitic effects. We have previously demonstrated that CHE can inhibit proliferation and promote apoptosis in human hepatocellular carcinoma (HCC) cells. However, the effect of CHE on the metastasis of HCC and its related molecular mechanisms have yet to be validated. In this study, we investigated the effects of CHE on the migration and invasion of the HCC cell line Hep3B. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), wounding healing, transwell migration and invasion assays and cytoskeleton staining demonstrated that CHE could inhibit the migration and invasion of Hep3B cells in a dose-dependent manner with change of cell structure. RNA interference studies made a knockdown of matrix metalloproteinase (MMP)-2/9 respectively in Hep3B cells. And the results of wounding healing and transwell invasion assay with the treatment of small interfering RNA (siRNA) investigated that MMP-2/9 are positively associated with Hep3B cell metastasis. The results of enzyme-linked immunosorbent assay (ELISA), Western blotting and quantitative RT-PCR showed that CHE suppressed the expression of MMP-2/9 at both mRNA and protein levels. CHE also exhibited an inhibitory effect on the phosphorylation of Focal adhesion kinase (FAK), phosphatidylinositol 3-kinase (PI3K), Akt, mammalian target of rapamycin (mTOR), c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinases (ERK) and p38. In summary, on Hep3B cells, CHE could change the cell cytoskeletal structures through reducing the expression of p-FAK and inhibit the metastasis of Hep3B cells by downregulating the expression of MMP-2/9 mainly through PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuanzhang Zhu
- Laboratory of Formula, School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Yingyi Pan
- Department of Traditional Chinese Medicine, School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Guibiao Zhang
- Department of Traditional Chinese Medicine, School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Yingchun Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Weicai Zhong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Chunxiao Chu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Yun Qian
- Laboratory of Formula, School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| | - Guofu Zhu
- Department of Traditional Chinese Medicine, School of Pharmacy, Shanghai University of Traditional Chinese Medicine
| |
Collapse
|
32
|
Chan KKS, Leung CON, Wong CCL, Ho DWH, Chok KSH, Lai CL, Ng IOL, Lo RCL. Secretory Stanniocalcin 1 promotes metastasis of hepatocellular carcinoma through activation of JNK signaling pathway. Cancer Lett 2017; 403:330-338. [PMID: 28688970 DOI: 10.1016/j.canlet.2017.06.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/19/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022]
Abstract
The hypoxic microenvironment is well-characterized in hepatocellular carcinoma (HCC). Delineation of hypoxia-responsive events is an integral part to understand the pathogenesis of HCC. We studied the functional role and clinical relevance of Stanniocalcin 1 (STC1), a hypoxia-induced molecular target, in HCC. In our clinical cohort, STC1 transcript was up-regulated in HCC tumor tissues. Moreover, STC1 protein was detected in the sera of HCC patients. A higher serum STC1 level was correlated with larger tumor size and poorer 5-year disease-free survival. Functionally, recombinant STC1 protein (rhSTC1) promoted cell migration and cell invasion in vitro; and the effect was abolished by co-treatment of anti-STC1 neutralizing antibody. By in vivo mouse model, silencing of STC1 in HCC cells downregulated secretory STC1 level and suppressed lung metastasis. Furthermore, we found that rhSTC1 activated the JNK pathway, as evidenced by altered expression of the key molecular targets pJNK and p-c-Jun. The functional effects conferred by rhSTC1 were abrogated by co-treatment of JNK inhibitor. In summary, secretory STC1 enhances metastatic potential of HCC via JNK signaling. It potentially serves as a prognostic serum biomarker and a therapeutic target for HCC.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/secondary
- Cell Movement/drug effects
- Disease-Free Survival
- Gene Expression Regulation, Neoplastic
- Glycoproteins/blood
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Humans
- JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors
- JNK Mitogen-Activated Protein Kinases/metabolism
- Kaplan-Meier Estimate
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Lung Neoplasms/enzymology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mice, Inbred BALB C
- Mice, Nude
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- RNA Interference
- Signal Transduction/drug effects
- Time Factors
- Transfection
- Tumor Burden
- Tumor Hypoxia
- Tumor Microenvironment
- Up-Regulation
Collapse
Affiliation(s)
- Kristy Kwan-Shuen Chan
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Carmen Oi-Ning Leung
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Carmen Chak-Lui Wong
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Daniel Wai-Hung Ho
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kenneth Siu-Ho Chok
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ching-Lung Lai
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Irene Oi-Lin Ng
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong
| | - Regina Cheuk-Lam Lo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
33
|
Zandberga E, Zayakin P, Ābols A, Pūpola D, Trapencieris P, Linē A. Depletion of carbonic anhydrase IX abrogates hypoxia-induced overexpression of stanniocalcin-1 in triple negative breast cancer cells. Cancer Biol Ther 2017; 18:596-605. [PMID: 28665755 DOI: 10.1080/15384047.2017.1345390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Carbonic anhydrase IX (CAIX) is a pH-regulating enzyme that plays a key role in maintaining an alkaline intracellular pH under hypoxic conditions. It is overexpressed in a variety of solid cancers, including breast cancer (BC), and has been implicated in the migration, invasion and stemness of breast cancer cells. Therefore, CAIX recently emerged as a novel therapeutic target for the treatment of BC. To gain an insight into the mechanism of action of CAIX inhibitors, we investigated the impact of CAIX knock-down on the transcriptional response to hypoxia in 2 BC cell lines - MCF7 and MDA-MB-231, by performing a global gene expression analysis. This showed that CAIX knock-down had a relatively minor effect on the global transcriptional response to hypoxia, however it blocked hypoxia-induced upregulation of stanniocalcin-1 (STC1), a secreted glycoprotein that has been shown to promote tumor progression and metastasis in BC. Kaplan-Meier survival analysis showed that high STC1 expression is significantly associated with poor survival in patients with basal-type breast cancer but not luminal A and HER2+ subtypes. Moreover, the association was particularly high in a subgroup of basal-type BC patients with TP53 mutations thus revealing a putative cooperation of STC1 with mutated TP53 in generating highly aggressive BC subgroup. Taken together, these findings show that CAIX inhibitors at least partially act through blocking STC1 induction in BC cells and reveal a subgroup of BC patients, who potentially would benefit most from the treatment with CAIX inhibitors.
Collapse
Affiliation(s)
- Elīna Zandberga
- a Latvian Biomedical Research and Study Centre , Riga , Latvia
| | - Pawel Zayakin
- a Latvian Biomedical Research and Study Centre , Riga , Latvia
| | - Artūrs Ābols
- a Latvian Biomedical Research and Study Centre , Riga , Latvia
| | - Dārta Pūpola
- a Latvian Biomedical Research and Study Centre , Riga , Latvia
| | | | - Aija Linē
- a Latvian Biomedical Research and Study Centre , Riga , Latvia.,c Faculty of Biology, University of Latvia , Riga , Latvia
| |
Collapse
|