1
|
Wang J, Sun Y, Wu R. ACSM3 Suppresses Ovarian Cancer Progression by Inactivating the IFN-γ/JAK/STAT3 Signaling Pathway. Adv Biol (Weinh) 2025; 9:e2400093. [PMID: 39913127 DOI: 10.1002/adbi.202400093] [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: 02/19/2024] [Revised: 11/12/2024] [Indexed: 02/07/2025]
Abstract
This study aimed to investigate the role of Acyl-CoA medium-chain synthetase 3 (ACSM3) in ovarian cancer (OC) progression. ACSM3 expression in OC tissues and cells is detected by real-time quantitative polymerase chain reaction, immunohistochemistry, or western blot. The influences of ACSM3 on OC progression are assessed in vitro and in vivo experiments. Gene set enrichment analysis is performed to find significantly enriched pathways related to ACSM3. In this study, ACSM3 expression in OC tissues and cells is downregulated. ACSM3 inhibited tumor growth in vivo. Knockdown of ACSM3 promoted cell proliferation, migration, and invasion, inhibited cell apoptosis, and activated JAK/STAT3 signaling pathway in SKOV3 cells, while overexpression of ACSM3 in A2780 cells led to the opposite results. Moreover, treatment with interferon-gamma (IFN-γ) in A2780 cells reversed the effects of ACSM3 on cell proliferation, migration, invasion, and apoptosis, but IFN-γ further enhanced the effects of ACSM3 knockdown on SKOV3 cell proliferation, migration, invasion, and apoptosis. In conclusion, ACSM3 inhibited OC cell proliferation, migration, and invasion, and promoted cell apoptosis by suppressing the IFN-γ/JAK/STAT3 signaling pathway, which might provide a promising therapeutic target for OC treatment.
Collapse
Affiliation(s)
- Juan Wang
- Department of Gynecology, Bin Zhou Medical University Hospital, Binzhou, Shandong, 256603, P. R. China
| | - Yanqiu Sun
- Department of obstetrics and gynecology, Bin Zhou Medical University Hospital, Binzhou, Shandong, 256603, P. R. China
| | - Ruixue Wu
- Department of Gynecology, Bin Zhou Medical University Hospital, Binzhou, Shandong, 256603, P. R. China
| |
Collapse
|
2
|
Karaman E, Yay F, Ayan D, Bayram E, Erturk S. The Clinopathological and Prognostic Significance of SPOCK1 in Gynecological Cancers: A Bioinformatics Based Analysis. BIOLOGY 2025; 14:209. [PMID: 40001977 PMCID: PMC11852031 DOI: 10.3390/biology14020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Background: Sparc/osteonectin, cwcv, and kazal-like domains proteoglycan 1 (SPOCK1) is an oncogene that promotes tumor formation and progression in certain types of cancer and is associated with poor survival rates. However, there is limited information on the importance of SPOCK1 in gynecological cancers in the literature. The aim of this study was to explore the role of SPOCK1 in ovarian serous cystadenocarcinoma (OV), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), and uterine corpus endometrial carcinomas (UCEC). Methods: The data used in this study were obtained from the GEPIA2, TCGA, Kaplan-Meier Plotter, GeneMANIA, UALCAN, cBioPortal, and TIMER databases. Overall survival (OS) and relapse-free survival (RFS) rates were evaluated by Kaplan-Meier survival analysis. Spearman's rho and statistical significance values were obtained for the correlation between SPOCK1 expression and tumor infiltration by different immune cells. Results: Lower SPOCK1 gene expression was observed in CESC and UCEC compared to normal tissue (p < 0.05), but the OV did not differ significantly (p > 0.05). In OV, SPOCK1 gene expression was solely linked to age; in CESC, it was linked to age, stage, weight, and histology; and in UCEC, it was linked to age, stage, weight, and menopausal status. Conclusions:SPOCK1 gene expression in UCEC showed weak positive correlations with CD8+ T cells and weak negative correlations with CD4+ T cells. SPOCK1 may be a potential prognostic and therapeutic target for gynecological cancers.
Collapse
Affiliation(s)
- Enes Karaman
- Department of Obstetrics and Gynecology, Faculty of Medicine, Nigde Omer Halisdemir University, 51240 Nigde, Turkey
| | - Fatih Yay
- Medical Biochemistry, Nigde Omer Halisdemir University Research and Training Hospital, 51100 Nigde, Turkey
| | - Durmus Ayan
- Department of Medical Biochemistry, Faculty of Medicine, Nigde Omer Halisdemir University, 51240 Nigde, Turkey
| | - Ergul Bayram
- Medical Biochemistry, Nigde Omer Halisdemir University Research and Training Hospital, 51100 Nigde, Turkey
| | - Sefa Erturk
- Department of Biophysics, Faculty of Medicine, Nigde Omer Halisdemir University, 51240 Nigde, Turkey
| |
Collapse
|
3
|
Zhang X, Zhang X, Yin H, Li Q, Fan B, Jiang B, Xie A, Guo D, Hao H, Zhang B. Roles of SPOCK1 in the Formation Mechanisms and Treatment of Non-Small-Cell Lung Cancer and Brain Metastases from Lung Cancer. Onco Targets Ther 2025; 18:35-47. [PMID: 39835273 PMCID: PMC11745074 DOI: 10.2147/ott.s483576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Lung cancer is a malignant tumor with high morbidity and mortality in China and worldwide. Once it metastasizes to the brain, its prognosis is very poor. Brain metastases are found in about 20% of newly diagnosed non-small-cell lung cancer (NSCLC) patients. About 30% of NSCLC patients develop brain metastases during treatment. NSCLC that is positive for EGFR, ALK, and ROS1 variations is especially likely to metastasize to the brain. SPOCK1 is a proteoglycan with systemic physiological functions. It regulates the self-renewal of brain metastasis-initiating cells, regulates invasion and metastasis from the lung to the brain, plays an important role in tumor progression and treatment resistance, and has higher expression in metastatic tumor tissues than other tissues. Current treatments for NSCLC brain metastases include surgery, whole-brain radiotherapy, stereotactic radiotherapy, targeted therapy, and chemotherapy. SPOCK1 is involved in many signaling pathways, by which it influences a variety of NSCLC treatment methods. In this paper, the progress of research on the treatment of NSCLC brain metastases is reviewed to guide decisions on treatment options in clinical practice.
Collapse
Affiliation(s)
- Xuebing Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Xia Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
- Department of Oncology, Dalian Fifth People’s Hospital, Dalian, Liaoning, People’s Republic of China
| | - Hang Yin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Qizheng Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Buqun Fan
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Bolun Jiang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Anqi Xie
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Dandan Guo
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Huanling Hao
- Department of Oncology, Dandong First Hospital, Dandong, Liaoning, People’s Republic of China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| |
Collapse
|
4
|
Xiao M, Xue J, Jin E. SPOCK: Master regulator of malignant tumors (Review). Mol Med Rep 2024; 30:231. [PMID: 39392048 PMCID: PMC11487499 DOI: 10.3892/mmr.2024.13355] [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: 05/27/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
SPARC/osteonectin, CWCV and Kazal‑like domain proteoglycan (SPOCK) is a family of highly conserved multidomain proteins. In total, three such family members, SPOCK1, SPOCK2 and SPOCK3, constitute the majority of extracellular matrix glycoproteins. The SPOCK gene family has been demonstrated to serve key roles in tumor regulation by affecting MMPs, which accelerates the progression of cancer epithelial‑mesenchymal transition. In addition, they can regulate the cell cycle via overexpression, inhibit tumor cell proliferation by inactivating PI3K/AKT signaling and have been associated with numerous microRNAs that influence the expression of downstream genes. Therefore, the SPOCK gene family are potential cancer‑regulating genes. The present review summarizes the molecular structure, tissue distribution and biological function of the SPOCK family of proteins, in addition to its association with cancer. Furthermore, the present review documents the progress made in investigations into the role of SPOCK, whilst also discussing prospects for the future of SPOCK‑targeted therapy, to provide novel ideas for clinical application and treatment.
Collapse
Affiliation(s)
- Mingyuan Xiao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| | - Jiancheng Xue
- Department of Otolaryngology, Head and Neck Surgery, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
- Shenzhen Clinical Research Center for Otolaryngology Diseases, The Second People's Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Enli Jin
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110134, P.R. China
| |
Collapse
|
5
|
Zhang T, Chen L, Xu X, Shen C. Knockdown of Long Noncoding RNA Urothelial Carcinoma-Associated 1 Represses Gallbladder Cancer Advancement by Regulating SPOCK1 Expression Through Sponging miR-613. Cancer Biother Radiopharm 2023; 38:354-363. [PMID: 33090888 DOI: 10.1089/cbr.2020.4290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Gallbladder cancer (GBC) is the most common biliary tract malignancy. Long noncoding RNA urothelial carcinoma-associated 1 (UCA1) and MicroRNA-613 (miR-613) have been reported to be involved in the progression of various cancers. However, the regulatory mechanism between UCA1 and miR-613 in GBC is unclear. Materials and Methods: The expression levels of UCA1, miR-613, and secreted protein/osteonectin, cwcv, and kazal-like domains proteoglycan 1 (SPOCK1) mRNA were detected using quantitative real-time polymerase chain reaction. Cell proliferation, migration, invasion, and apoptosis were determined with MTT, transwell, or flow cytometry assays. The levels of SPOCK1 protein, Bax, cleaved-casp-3, and Bcl-2 were determined by Western blot analysis. The relationship between miR-613 and UCA1 or SPOCK1 was verified through dual-luciferase reporter and/or RNA immunoprecipitation assays. Xenograft assay was performed to verify the role of UCA1 in vivo. Results: UCA1 and SPOCK1 were upregulated, whereas miR-613 was downregulated in GBC tissues and cells. UCA1 silencing decreased tumor growth in vivo and impeded proliferation, migration, invasion, and induced apoptosis of GBC cells in vitro. Notably, UCA1 acted as a sponge for miR-613, which targeted SPOCK1 in GBC cells. Moreover, UCA1 enhancement reversed the repressive impact of miR-613 mimic on the malignancy of GBC cells. UCA1 regulated SPOCK1 expression through adsorbing miR-613. Furthermore, SPOCK1 elevation overturned UCA1 silencing mediated the malignant behaviors of GBC cells. Conclusion: UCA1 knockdown suppressed GBC progression through downregulating SPOCK1 via sponging miR-613, providing an evidence for UCA1 as a target for GBC treatment.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Hepatobiliary Surgery, Loudi Central Hospital of Hunan, Loudi, China
| | - Lijian Chen
- Department of General Surgery, Hunan Children's Hospital, Changsha, China
| | - Xundi Xu
- Department of Hepatobiliary Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chao Shen
- Department of Urology, Loudi Central Hospital of Hunan, Loudi, China
| |
Collapse
|
6
|
Váncza L, Horváth A, Seungyeon L, Rókusz A, Dezső K, Reszegi A, Petővári G, Götte M, Kovalszky I, Baghy K. SPOCK1 Overexpression Suggests Poor Prognosis of Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15072037. [PMID: 37046698 PMCID: PMC10093273 DOI: 10.3390/cancers15072037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/20/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023] Open
Abstract
Purpose: Sparc/osteonectin, cwcv, and kazal-like domains proteoglycan 1 (SPOCK1) has been found in a variety of malignant tumors and is associated with a poor prognosis. We aimed to explore the role of SPOCK1 in ovarian cancer. Methods: Ovarian cancer cell lines SKOV3 and SW626 were transfected with SPOCK1 overexpressing or empty vector using electroporation. Cells were studied by immunostaining and an automated Western blotting system. BrdU uptake and wound healing assays assessed cell proliferation and migration. SPOCK1 expression in human ovarian cancer tissues and in blood samples were studied by immunostaining and ELISA. Survival of patients with tumors exhibiting low and high SPOCK1 expression was analyzed using online tools. Results: Both transfected cell lines synthesized different SPOCK1 variants; SKOV3 cells also secreted the proteoglycan. SPOCK1 overexpression stimulated DNA synthesis and cell migration involving p21CIP1. Ovarian cancer patients had increased SPOCK1 serum levels compared to healthy controls. Tumor cells of tissues also displayed abundant SPOCK1. Moreover, SPOCK1 levels were higher in untreated ovarian cancer serum and tissue samples and lower in recipients of chemotherapy. According to in silico analyses, high SPOCK1 expression was correlated with shorter survival. Conclusion: Our findings suggest SPOCK1 may be a viable anti-tumor therapeutic target and could be used for monitoring ovarian cancer.
Collapse
|
7
|
Lin YW, Wen YC, Hsiao CH, Lai FR, Yang SF, Yang YC, Ho KH, Hsieh FK, Hsiao M, Lee WJ, Chien MH. Proteoglycan SPOCK1 as a Poor Prognostic Marker Promotes Malignant Progression of Clear Cell Renal Cell Carcinoma via Triggering the Snail/Slug-MMP-2 Axis-Mediated Epithelial-to-Mesenchymal Transition. Cells 2023; 12:cells12030352. [PMID: 36766694 PMCID: PMC9913795 DOI: 10.3390/cells12030352] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Sparc/osteonectin, cwcv, and kazal-like domains proteoglycan 1 (SPOCK1) has been reported to play an oncogenic role in certain cancer types; however, the role of SPOCK1 in the progression of clear cell renal cell carcinoma (ccRCC) remains elusive. Here, higher SPOCK1 transcript and protein levels were observed in ccRCC tissues compared to normal tissues and correlated with advanced clinical stages, larger tumor sizes, and lymph node and distal metastases. Knockdown and overexpression of SPOCK1 in ccRCC cells led to decreased and increased cell clonogenic and migratory/invasive abilities in vitro as well as lower and higher tumor growth and invasion in vivo, respectively. Mechanistically, the gene set enrichment analysis (GSEA) database was used to identify the gene set of epithelial-to-mesenchymal transition (EMT) pathways enriched in ccRCC samples with high SPOCK1 expression. Further mechanistic investigations revealed that SPOCK1 triggered the Snail/Slug-matrix metalloproteinase (MMP)-2 axis to promote EMT and cell motility. Clinical ccRCC samples revealed SPOCK1 to be an independent prognostic factor for overall survival (OS), and positive correlations of SPOCK1 with MMP-2 and mesenchymal-related gene expression levels were found. We observed that patients with SPOCK1high/MMP2high tumors had the shortest OS times compared to others. In conclusion, our findings reveal that SPOCK1 can serve as a useful biomarker for predicting ccRCC progression and prognosis, and as a promising target for treating ccRCC.
Collapse
Affiliation(s)
- Yung-Wei Lin
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 11031, Taiwan
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yu-Ching Wen
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 11031, Taiwan
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chi-Hao Hsiao
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 11031, Taiwan
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Feng-Ru Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 404, Taiwan
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Research, Tungs’ Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
| | - Kuo-Hao Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Feng-Koo Hsieh
- The Genome Engineering & Stem Cell Center, School of Medicine, Washington University, St. Louis, MO 63105, USA
| | - Michael Hsiao
- The Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wei-Jiunn Lee
- Department of Urology, School of Medicine, College of Medicine and TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Correspondence: (W.-J.L.); (M.-H.C.); Tel.: +886-2-27-361-661 (ext. 3237) (M.-H.C.); Fax: +886-2-27-390-500 (M.-H.C.)
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Correspondence: (W.-J.L.); (M.-H.C.); Tel.: +886-2-27-361-661 (ext. 3237) (M.-H.C.); Fax: +886-2-27-390-500 (M.-H.C.)
| |
Collapse
|
8
|
Paskaš S, Murganić B, Kuhnert R, Hey-Hawkins E, Mijatović S, Maksimović-Ivanić D. Carborane-Based Analog of Rev-5901 Attenuates Growth of Colon Carcinoma In Vivo. Molecules 2022; 27:molecules27144503. [PMID: 35889376 PMCID: PMC9321230 DOI: 10.3390/molecules27144503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Lipoxygenases convert polyunsaturated fatty acids into biologically active metabolites such as inflammatory mediators—prostaglandins and leukotrienes. The inhibition of lipoxygenases is increasingly employed in the treatment of cancer. We evaluated the anticancer potential of two novel 5-lipoxygenase inhibitors, named CarbZDNaph and CarbZDChin, which are analogues of the commercially available inhibitor Rev-5901. The in vitro segment of this study was conducted on a mouse colorectal carcinoma cell line—CT26CL25. For an in vivo model, we induced tumors in BALB/c mice by the implantation of CT26CL25 cells, and we treated the animals with potential inhibitors. A 48 h treatment resulted in diminished cell viability. Calculated IC50 values (half-maximal inhibitory concentrations) were 25 μM, 15 μM and 30 μM for CarbZDNaph, CarbZDChin and Rev-5901, respectively. The detailed analysis of mechanism revealed an induction of caspase-dependent apoptosis and autophagy. In the presence of chloroquine, an autophagy inhibitor, we observed an increased mortality of cells, implying a cytoprotective role of autophagy. Our in vivo experiment reports tumor growth attenuation in animals treated with CarbZDChin. Compounds CarbZDNaph and Rev-5901 lacked an in vivo efficacy. The results presented in this study display a strong effect of compound CarbZDChin on malignant cell growth. Having in mind the important role of inflammation in cancer development, these results have a significant impact and are worthy of further evaluation.
Collapse
Affiliation(s)
- Svetlana Paskaš
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
| | - Blagoje Murganić
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
| | - Robert Kuhnert
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany; (R.K.); (E.H.-H.)
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany; (R.K.); (E.H.-H.)
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
- Correspondence: ; Tel.: +381-11-2078452
| |
Collapse
|
9
|
SPOCK1 silencing decreases 5-FU resistance through PRRX1 in colorectal cancer. Pathol Res Pract 2022; 234:153895. [DOI: 10.1016/j.prp.2022.153895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/25/2022] [Accepted: 04/09/2022] [Indexed: 11/19/2022]
|
10
|
Luo Y, Liu W, Zhu Y, Tian Y, Wu K, Ji L, Ding L, Zhang W, Gao T, Liu X, Zhao J. KIF11 as a potential cancer prognostic marker promotes tumorigenesis in children with Wilms tumor. Pediatr Hematol Oncol 2022; 39:145-157. [PMID: 34378481 DOI: 10.1080/08880018.2021.1953655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Emerging evidence suggests that KIF11 could play a pivotal role in cancer cell proliferation; however, its biological functions and molecular mechanisms in Wilms tumor (WT) cells are largely unknown. The aim of this study was to evaluate the clinical significance and therapeutic potential of KIF11 proteins in WT. KIF11 expression in WT tissues and adjacent nontumor tissues was determined using qRT-PCR, Western blotting, immunohistochemistry (IHC) and bioinformatics. The function of KIF11 protein was determined by its correlation with tumor cell growth, angiogenesis, and apoptosis using IHC and lentiviral vector-mediated KIF11 depletion. KIF11 expression was upregulated in WT tissues and was associated with WT clinical outcomes. Tumor KIF11 expression was significantly associated with the Ki67 proliferation index. CCK-8, flow-cytometric analysis, and Western blotting revealed that KIF11 knockdown significantly inhibited WT cell growth. Functional studies have indicated that increased KIF11 expression is significantly correlated with vascular endothelial growth factor (VEGF) expression and intratumoral microvessel density. We further confirmed that downregulated expression of KIF11 promoted cell apoptosis and significantly increased Bcl-2 and Bax expression. Our findings demonstrate that KIF11 plays a role in promoting the development of human WT and can serve as a potential molecular marker for the treatment of WT.
Collapse
Affiliation(s)
- Yishu Luo
- School of Medicine, Nantong University, Nantong, China
| | - Wei Liu
- Department of General Surgery, Yancheng Third People's Hospital, Yancheng, China
| | - Yinmei Zhu
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongshen Tian
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Ke Wu
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Linghua Ji
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Li Ding
- School of Medicine, Nantong University, Nantong, China
| | - Wenwen Zhang
- Department of Radiation Oncology, Nantong Third People's Hospital, Nantong, China
| | - Tingting Gao
- Department of General Surgery, Shanghai children's Hospital, Shanghai, China
| | - Xiaoqin Liu
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jun Zhao
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
11
|
Liu Y, Zhu X, Zhang W, Bian T, Wu Z, Zhang J, Qiu H, Hu Y, Feng J, Shi J. RASSF10 exhibits tumor‑suppressing potential involving tumor proliferation, metastasis and epithelial‑mesenchymal transition in esophageal squamous cell carcinoma. Oncol Rep 2022; 47:80. [PMID: 35211758 PMCID: PMC8892611 DOI: 10.3892/or.2022.8291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
Growing evidence indicates that Ras-association domain family 10 (RASSF10) is a novel tumor-suppressor gene that is involved in the inhibition of tumor progression and metastasis; however, the biological functions and molecular mechanisms of RASSF10 in esophageal squamous cell carcinoma (ESCC) have not yet been thoroughly elucidated. The expression of RASSF10 in ESCC tissues and adjacent non-tumor tissues was investigated employing quantitative polymerase chain reaction (qPCR) and immunohistochemistry (IHC) assays of tissue microarrays. The function of RASSF10 in ESCC cell growth, migration and invasion was determined by CCK-8, colony formation, scratch wound healing and Transwell invasion assays, respectively. The correlation between RASSF10 and markers related to epithelial-mesenchymal transition (EMT) was evaluated by tissue microarray (TMA)-IHC, western blotting and immunofluorescence staining. RASSF10 was found to be highly downregulated in ESCC tissues compared with that noted in the adjacent non-tumor tissues, and closely correlated with tumor progression and patient prognosis. Moreover, functional studies demonstrated that RASSF10 overexpression not only resulted in reduced cell growth and colony formation but also inhibited migration and invasion of the ESCC cells. Tumor RASSF10 expression was positively correlated with E-cadherin expression and negatively correlated with vimentin. In addition, it was demonstrated that the antineoplastic functions of RASSF10 mediate inactivation of the Wnt/β-catenin pathway in ESCC. Our findings revealed that RASSF10 may constitute a prognostic factor for ESCC patients and a crucial candidate for targeted therapy against ESCC.
Collapse
Affiliation(s)
- Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Xiaohui Zhu
- Department of Internal Medicine, Nantong Tumor Hospital, Nantong, Jiangsu 226600, P.R. China
| | - Wenwen Zhang
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Zheng Wu
- Department of Respiration, Affiliated Hai'an Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Hongmei Qiu
- Department of Respiration, Nantong Geriatric Rehabilitation Hospital, Branch of the Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Yingzi Hu
- Medical School of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Jia Feng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| | - Jiahai Shi
- Department of Cardio‑Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226600, P.R. China
| |
Collapse
|
12
|
Babaei-Jadidi R, Kashfi H, Alelwani W, Karimi Bakhtiari A, Kattan SW, Mansouri OA, Mukherjee A, Lobo DN, Nateri AS. Anti-miR-135/SPOCK1 axis antagonizes the influence of metabolism on drug response in intestinal/colon tumour organoids. Oncogenesis 2022; 11:4. [PMID: 35046388 PMCID: PMC8770633 DOI: 10.1038/s41389-021-00376-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Little is known about the role of microRNAs (miRNAs) in rewiring the metabolism within tumours and adjacent non-tumour bearing normal tissue and their potential in cancer therapy. This study aimed to investigate the relationship between deregulated miRNAs and metabolic components in murine duodenal polyps and non-polyp-derived organoids (mPOs and mNPOs) from a double-mutant ApcMinFbxw7∆G mouse model of intestinal/colorectal cancer (CRC). We analysed the expression of 373 miRNAs and 12 deregulated metabolic genes in mPOs and mNPOs. Our findings revealed miR-135b might target Spock1. Upregulation of SPOCK1 correlated with advanced stages of CRCs. Knockdown of miR-135b decreased the expression level of SPOCK1, glucose consumption and lactic secretion in CRC patient-derived tumours organoids (CRC tPDOs). Increased SPOCK1 induced by miR-135b overexpression promoted the Warburg effect and consequently antitumour effect of 5-fluorouracil. Thus, combination with miR-135b antisense nucleotides may represent a novel strategy to sensitise CRC to the chemo-reagent based treatment.
Collapse
Affiliation(s)
- Roya Babaei-Jadidi
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
- Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Hossein Kashfi
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Walla Alelwani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ashkan Karimi Bakhtiari
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Shahad W Kattan
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
- Medical Laboratory Department, College of Applied Medical Sciences, Taibah University, Yanbu, Saudi Arabia
| | - Omniah A Mansouri
- Department of Biology, University of Jeddah, College of Science, Jeddah, 21959, Saudi Arabia
| | - Abhik Mukherjee
- Histopathology, BioDiscovery Institute, School of Medicine, University of Nottingham, NG7 2UH, Nottingham, UK
| | - Dileep N Lobo
- Nottingham Digestive Diseases Centre, National Nottingham Digestive Diseases Centre, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Abdolrahman S Nateri
- Cancer Genetics & Stem Cell Group, BioDiscovery Institute, Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
13
|
Han J, Rong Y, Gao X. Multiomic analysis of the function of SPOCK1 across cancers: an integrated bioinformatics approach. J Int Med Res 2021; 49:300060520962659. [PMID: 34156309 PMCID: PMC8236807 DOI: 10.1177/0300060520962659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective To investigate SPARC (osteonectin), cwcv and kazal like domains proteoglycan 1 (SPOCK1) gene expression across The Cancer Genome Atlas (TCGA) cancers, both in cancer versus normal tissues and in different stages across the cancer types. Methods This integrated bioinformatics study used data from several bioinformatics databases (Cancer Cell Line Encyclopedia, Genotype-Tissue Expression, TCGA, Tumor Immune Estimation Resource [TIMER]) to define the expression pattern of the SPOCK1 gene. A survival analysis was undertaken across the cancers. The search tool for retrieval of interacting genes (STRING) database was used to identify proteins that interacted with SPOCK1. Gene Set Enrichment Analysis was conducted to determine pathway enrichment. The TIMER database was used to explore the correlation between SPOCK1 and immune cell infiltration. Results This multiomic analysis showed that the SPOCK1 gene was expressed differently between normal tissues and tumours in several cancers and that it was involved in cancer progression. The overexpression of the SPOCK1 gene was associated with poor clinical outcomes. Analysis of gene expression and tumour-infiltrating immune cells showed that SPOCK1 correlated with several immune cells across cancers. Conclusions This research showed that SPOCK1 might serve as a new target for several cancer therapies in the future.
Collapse
Affiliation(s)
- Jie Han
- Department of Hepatology, Qilu Hospital, Shandong University, Shandong, China
| | - Yihui Rong
- Infection Disease Center of Peking University International Hospital, Beijing, China
| | - Xudong Gao
- Infection Disease Center of Peking University International Hospital, Beijing, China
| |
Collapse
|
14
|
Wang B, Tang D, Liu Z, Wang Q, Xue S, Zhao Z, Feng D, Sheng C, Li J, Zhou Z. LINC00958 promotes proliferation, migration, invasion, and epithelial-mesenchymal transition of oesophageal squamous cell carcinoma cells. PLoS One 2021; 16:e0251797. [PMID: 34003875 PMCID: PMC8130937 DOI: 10.1371/journal.pone.0251797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/03/2021] [Indexed: 02/03/2023] Open
Abstract
Oesophageal cancer is one of the deadliest cancers in the world. Oesophageal squamous cell carcinoma (ESCC) is the most prevalent histological type of oesophageal cancer. Oesophageal cancer has a poor prognosis because of its invasiveness. Thus, it is especially important to seek effective treatment methods. Research indicates that long non-coding RNAs (lncRNAs) play a significant role in the occurrence and development of oesophageal cancer. The aim of this study was to describe the role of LINC00958 in ESCC. Bioinformatics and real-time quantitative polymerase chain reaction (RT-qPCR) methods were utilized to predict and verify the expression of LINC00958 in ESCC. Related functional experiments, including cell proliferation, migration and invasion, were performed. In addition, a western blot and a dual luciferase reporter gene experiment were used to study the detailed carcinogenic mechanism of LINC00958. The results indicated there was a high expression of LINC00958 in ESCC, which promoted proliferation, migration, invasion and Epithelial–Mesenchymal Transition (EMT) of ESCC cells, and this effect may be via regulating miR-510-5p.
Collapse
Affiliation(s)
- Biqi Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Duo Tang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Zijia Liu
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Qian Wang
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Shan Xue
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Zijie Zhao
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Dongdong Feng
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Chao Sheng
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Jintao Li
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
| | - Zhixiang Zhou
- Beijing International Science and Technology Cooperation Base of Antivirus Drug, Faculty of environment and life, Beijing University of Technology, Beijing, China
- * E-mail:
| |
Collapse
|
15
|
Shan Z, Wu W, Yan X, Yang Y, Luo D, Liu Q, Li X, Goel A, Ma Y. A novel epithelial-mesenchymal transition molecular signature predicts the oncological outcomes in colorectal cancer. J Cell Mol Med 2021; 25:3194-3204. [PMID: 33660944 PMCID: PMC8034457 DOI: 10.1111/jcmm.16387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
Epithelial‐mesenchymal transition (EMT), a biological process involving the transformation of epithelial cells into mesenchymal cells, promotes tumour initiation and metastasis. The aim of this study was to construct an EMT molecular signature for predicting colorectal cancer (CRC) prognosis and evaluate the efficacy of the model. The risk scoring system, constructed by log‐rank test and multivariate Cox regression analysis according to EMT‐related gene expression in CRC patients from TCGA database, demonstrated the highest correlation with prognosis compared with other parameters in CRC patients. The risk scores were significantly correlated with more lymph node metastasis, distal metastasis and advanced clinical stage of CRC. The model was further successfully validated in two independent external cohorts from GEO database. Furthermore, we developed a nomogram to integrate the EMT signature with the pathological stage of CRC, which was found to perform well in predicting the overall survival. Additionally, this risk scoring model was found to be associated with immune cell infiltration, implying a potential role of EMT involved in immunity regulation in tumour microenvironment. Taken together, our novel EMT molecular model may be useful in identifying high‐risk patients who need an intensive follow‐up and more aggressive therapy, finally contributing to more precise individualized therapeutic strategies.
Collapse
Affiliation(s)
- Zezhi Shan
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen Wu
- Department of Surgery, Shanghai Pudong Hospital (Fudan University Pudong Medical Center), Shanghai, China
| | - Xuebing Yan
- Department of Oncology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dakui Luo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Zhu X, Jiang S, Wu Z, Liu T, Zhang W, Wu L, Xu L, Shao M. Long non-coding RNA TTN antisense RNA 1 facilitates hepatocellular carcinoma progression via regulating miR-139-5p/SPOCK1 axis. Bioengineered 2021; 12:578-588. [PMID: 33517826 PMCID: PMC8291788 DOI: 10.1080/21655979.2021.1882133] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Reportedly, long non-coding RNAs (lncRNAs) are implicated in hepatocellular carcinoma (HCC) progression, yet little is known concerning the biological functions of TTN antisense RNA 1 (TTN-AS1) in HCC. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) was performed for detecting TTN-AS1, SPOCK1 mRNA, and miR-139-5p expressions in HCC cells and tissues. After TTN-AS1 was overexpressed or knocked down in HCC cells, CCK-8 and 5-Ethynyl-2ʹ-deoxyuridine (EdU) assays were carried out for examining cell multiplication. Transwell assays were conducted for evaluating HCC cell migration and invasion. Dual-luciferase reporter assay was employed for verifying the binding relationships between miR-139-5p and TTN-AS1, and between SPOCK1 3ʹUTR and miR-139-5p. Western blot was employed to measure SPOCK1, E-cadherin, N-cadherin, and Vimentin protein expressions. We demonstrated that, TTN-AS1 and SPOCK1 expression levels were remarkably enhanced in HCC cells and tissues, whereas miR-139-5p expression was observably reduced. Functional experiments suggested that TTN-AS1 knockdown markedly repressed HCC cell multiplication, migration, epithelial-mesenchymal transition (EMT), and invasion. In addition, TTN-AS1 interacted with miR-139-5p and decreased its expression. Moreover, SPOCK1 was a miR-139-5p target, and miR-139-5p inhibitors were able to reverse TTN-AS1 knockdown-induced inhibitory effect on SPOCK1 expression. SPOCK1 overexpression plasmid could counteract TTN-AS1 knockdown-induced inhibiting impact on HCC cell multiplication, migration, invasion, and EMT. In conclusion, TTN-AS1 expression level is remarkably enhanced in HCC, and TTN-AS1 can promote the multiplication, migration, invasion, and EMT of HCC cells via regulating miR-139-5p/SPOCK1 axis.
Collapse
Affiliation(s)
- Xinghao Zhu
- Department of Internal Medicine of Chinese Medicine, Henan University of Chinese Medicine , Zhengzhou, Henan, China
| | - Shiqing Jiang
- The First Affiliated Hospital of Henan University of Chinese Medicine , Zhengzhou, Henan, China
| | - Zongyao Wu
- Institute of Tibetan Medicine, Tibet University of Tibetan Medicine , Lhasa, Xizang, China
| | - Tonghua Liu
- Beijing University of Chinese Medicine , Beijing, China
| | - Wei Zhang
- Institute of Liver Diseases, Shijiazhuang Fifth Hospital , Shijiazhuang, Hebei, China
| | - Lili Wu
- Beijing University of Chinese Medicine , Beijing, China
| | - Lijun Xu
- Institute of Tibetan Medicine, Tibet University of Tibetan Medicine , Lhasa, Xizang, China
| | - Mingliang Shao
- Department of Oncology, Shijiazhuang Fifth Hospital , Shijiazhuang, Hebei, China
| |
Collapse
|
17
|
Xu M, Zhang X, Zhang S, Piao J, Yang Y, Wang X, Lin Z. SPOCK1/SIX1axis promotes breast cancer progression by activating AKT/mTOR signaling. Aging (Albany NY) 2020; 13:1032-1050. [PMID: 33293473 PMCID: PMC7835061 DOI: 10.18632/aging.202231] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 09/28/2020] [Indexed: 12/21/2022]
Abstract
SPOCK1 is highly expressed in many types of cancer and has been recognized as a promoter of cancer progression. Its regulatory mechanism in breast cancer (BC) remains unclear. This study aimed to explore the precise function of SPOCK1 in BC progression and to identify the mechanism by which SPOCK1 is involved in cell proliferation and epithelial-mesenchymal transition (EMT). Immunohistochemistry (IHC) experiments and database analysis showed that high expression of SPOCK1 was positively associated with histological grade, lymph node metastasis (LN) and poor clinical prognosis in BC. A series of in vitro and in vivo assays elucidated that altering the SPOCK1 level led to distinct changes in BC cell proliferation and metastasis. Investigations of potential mechanisms revealed that SPOCK1 interacted with SIX1 to enhance cell proliferation, cell cycle progression and EMT by activating the AKT/mTOR pathway, whereas inhibition of the AKT/mTOR pathway or depletion of SIX1 reversed the effects of SPOCK1 overexpression. Furthermore, SPOCK1 and SIX1 were highly expressed in BC and might indicate poor prognoses. Altogether, the SPOCK1/SIX1 axis promoted BC progression by activating the AKT/mTOR pathway to accelerate cell proliferation and promote metastasis in BC, so the SPOCK1/SIX1 axis might be a promising clinical therapeutic target for preventing BC progression.
Collapse
Affiliation(s)
- Ming Xu
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, South Korea
| | - Songnan Zhang
- Department of Oncology, Yanbian University Affiliated Hospital, Yanji, China
| | - Junjie Piao
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Yang Yang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Xinyue Wang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Zhenhua Lin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China
| |
Collapse
|
18
|
Zamanpoor M, Ghaedi H, Omrani MD. Association of SPOCK1 gene locus with rheumatoid arthritis in Caucasians. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
19
|
Colorectal Cancer Early Detection in Stool Samples Tracing CpG Islands Methylation Alterations Affecting Gene Expression. Int J Mol Sci 2020; 21:ijms21124494. [PMID: 32599859 PMCID: PMC7349989 DOI: 10.3390/ijms21124494] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/14/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer mortality. Early diagnosis is relevant for its prevention and treatment. Since DNA methylation alterations are early events in tumourigenesis and can be detected in cell-free DNA, they represent promising biomarkers for early CRC diagnosis through non-invasive methods. In our previous work, we identified 74 early altered CpG islands (CGIs) associated with genes involved in cell cross-talking and cell signalling pathways. The aim of this work was to test whether methylation-based biomarkers could be detected in non-invasive matrices. Our results confirmed methylation alterations of GRIA4 and VIPR2 in CRC tissues, using MethyLight, as well as in stool samples, using a much more sensitive technique as droplet digital PCR. Furthermore, we analysed expression levels of selected genes whose promoter CGIs were hypermethylated in CRC, detecting downregulation at mRNA and protein levels in CRC tissue for GRIA4, VIPR2, SPOCK1 and SLC6A3. Most of these genes were already lowly expressed in colon normal tissues supporting the idea that cancer DNA methylation targets genes already barely expressed in the matched normal tissues. Our study suggests GRIA4 and VIPR2 as biomarkers for early CRC diagnosis using stool samples and confirms downregulation of genes hypermethylated in CRC.
Collapse
|
20
|
SPOCK1 is a novel inducer of epithelial to mesenchymal transition in drug-induced gingival overgrowth. Sci Rep 2020; 10:9785. [PMID: 32555336 PMCID: PMC7300011 DOI: 10.1038/s41598-020-66660-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/22/2020] [Indexed: 11/28/2022] Open
Abstract
Few studies have investigated the role of extracellular-matrix proteoglycans in the pathogenesis of drug-induced gingival overgrowth (DIGO). SPOCK1 is an extracellular proteoglycan that induces epithelial to mesenchymal transition (EMT) in several cancer cell lines and exhibits protease-inhibitory activity. However, the role of SPOCK1 in non-cancerous diseases such as DIGO has not been well-addressed. We demonstrated that the expression of SPOCK1, TGF-β1, and MMP-9 in calcium channel blocker-induced gingival overgrowth is higher than that in non-overgrowth tissues. Transgenic mice overexpressing Spock1 developed obvious gingival-overgrowth and fibrosis phenotypes, and positively correlated with EMT-like changes. Furthermore, in vitro data indicated a tri-directional interaction between SPOCK1, TGF-β1, and MMP-9 that led to gingival overgrowth. Our study shows that SPOCK1 up-regulation in a noncancerous disease and SPOCK1-induced EMT in gingival overgrowth occurs via cooperation and crosstalk between several potential signaling pathways. Therefore, SPOCK1 is a novel therapeutic target for gingival overgrowth and its expression is a potential risk of EMT induction in cancerous lesions.
Collapse
|
21
|
Sun LR, Li SY, Guo QS, Zhou W, Zhang HM. SPOCK1 Involvement in Epithelial-to-Mesenchymal Transition: A New Target in Cancer Therapy? Cancer Manag Res 2020; 12:3561-3569. [PMID: 32547193 PMCID: PMC7244346 DOI: 10.2147/cmar.s249754] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/24/2022] Open
Abstract
Background Cancer metastasis is the main obstacle to increasing the lifespan of cancer patients. Epithelial-to-mesenchymal transition (EMT) plays a significant role in oncogenic processes, including tumor invasion, intravasation, and micrometastasis formation, and is especially critical for cancer invasion and metastasis. The extracellular matrix (ECM) plays a crucial role in the occurrence of EMT corresponding to the change in adhesion between cells and matrices. Conclusion SPOCK1 is a critical regulator of the ECM and mediates EMT in cancer cells. This suggests an important role for SPOCK1 in tumorigenesis, migration and invasion. SPOCK1 is a critical regulator of some processes involved in cancer progression, including cancer cell proliferation, apoptosis and migration. Herein, the functions of SPOCK1 in cancer progression are expounded, revealing the association between SPOCK1 and EMT in cancer metastasis. SPOCK1 is a positive downstream regulator of transforming growth factor-β, and SPOCK1-mediated EMT regulates invasion and metastasis through the Wnt/β-catenin pathway and PI3K/Akt signaling pathway. It is of significance that SPOCK1 may be an attractive prognostic biomarker and therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Li-Rui Sun
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Si-Yu Li
- Department of Pathology, Hangzhou Third Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Qiu-Shi Guo
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Wei Zhou
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Hong-Mei Zhang
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
22
|
Jiang W, Kai J, Li D, Wei Z, Wang Y, Wang W. lncRNA HOXB-AS3 exacerbates proliferation, migration, and invasion of lung cancer via activating the PI3K-AKT pathway. J Cell Physiol 2020; 235:7194-7203. [PMID: 32039488 DOI: 10.1002/jcp.29618] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022]
Abstract
Lung cancer remains the leading cause of cancer-related death all over the world. In spite of the great advances made in surgery and chemotherapy, the prognosis of lung cancer patients is poor. A substantial fraction of long noncoding RNAs (lncRNAs) can regulate various cancers. A recent study has reported that lncRNA HOXB-AS3 plays a critical role in cancers. However, its biological function remains unclear in lung cancer progression. In the current research, we found HOXB-AS3 was obviously elevated in NSCLC tissues and cells. Functional assays showed that inhibition of HOXB-AS3 was able to repress A549 and H1975 cell proliferation, cell colony formation ability and meanwhile, triggered cell apoptosis. Furthermore, the lung cancer cell cycle was mostly blocked in the G1 phase whereas the cell ratio in the S phase was reduced. Also, A549 and H1975 cell migration and invasion capacity were significantly repressed by the loss of HOXB-AS3. The PI3K/AKT pathway has been implicated in the carcinogenesis of multiple cancers. Here, we displayed that inhibition of HOXB-AS3 suppressed lung cancer cell progression via inactivating the PI3K/AKT pathway. Subsequently, in vivo experiments were utilized in our study and it was demonstrated that HOXB-AS3 contributed to lung cancer tumor growth via modulating the PI3K/AKT pathway. Overall, we implied that HOXB-AS3 might provide a new perspective for lung cancer treatment via targeting PI3K/AKT.
Collapse
Affiliation(s)
- Wenyang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jindan Kai
- Department of Thoracic Surgery, Hubei Cancer Hospital, Wuhan, China
| | - Donghang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongheng Wei
- Department of Oncology, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi, China
| | - Ying Wang
- Department of Respiratory Medicine, The First People's Hospital of Tianmen, Tianmen, Hubei, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
23
|
CLEC5A promotes the proliferation of gastric cancer cells by activating the PI3K/AKT/mTOR pathway. Biochem Biophys Res Commun 2020; 524:656-662. [PMID: 32033754 DOI: 10.1016/j.bbrc.2019.10.122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 02/08/2023]
Abstract
Gastric cancer (GC), as one of the most prevalent malignancies, contributes to the high morbidity and mortality worldwide. By analyzing the bioinformatics, qRT-PCR and IHC assays, we found that CLEC5A is overexpressed in GC and associated with poorer prognosis. CLEC5A silencing inhibits cell growth and DNA replication and induces cell cycle arrest and cell apoptosis. Bioinformatics analyses and Western blotting revealed that CLEC5A depletion led to the dysregulation of the PI3K/AKT/mTOR pathway. CLEC5A-mediated GC proliferation and anti-apoptosis were impaired by blocking the PI3K/AKT/mTOR pathway with LY294002. We hypothesize that CLEC5A is of vital importance to GC initiation and progression via the PI3K/AKT/mTOR pathway, and that our results might represent promising therapeutic strategies for GC patients.
Collapse
|
24
|
Ni QF, Zhang Y, Yu JW, Hua RH, Wang QH, Zhu JW. miR-92b promotes gastric cancer growth by activating the DAB2IP-mediated PI3K/AKT signalling pathway. Cell Prolif 2019; 53:e12630. [PMID: 31713929 PMCID: PMC6985694 DOI: 10.1111/cpr.12630] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/02/2019] [Accepted: 03/29/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives miR‐92b has been reported to play critical roles in several carcinomas; however, our understanding of the mechanisms by which miR‐92b stimulates gastric cancer (GC) is incomplete. The aim of this study was to investigate the clinical significance and functional relevance of miR‐92b in GC. Materials and methods Expression of miR‐92b in GC and peritumoural tissues was determined using qRT‐PCR, in situ hybridization and bioinformatics. CCK‐8, colony formation and fluorescence‐activated cell sorting assays were utilized to explore the effect of miR‐92b on GC cells. A luciferase reporter assay and Western blotting were employed to verify miR‐92b targeting of DAB2IP. Furthermore, Western blotting was used to evaluate the levels of DAB2IP and PI3K/Akt signalling pathway‐related proteins. Results In this study, we found that miR‐92b was upregulated in GC tissues compared with peritumoural tissues. Overexpression of miR‐92b promoted cell proliferation, colony formation, and G0/G1 transition and decreased apoptosis. Our results indicated that miR‐92b repressed the expression of DAB2IP and that loss of DAB2IP activated the PI3K/AKT signalling pathway. Overexpression of DAB2IP rescued the effects of miR‐92b in GC cells. Finally, our results demonstrated a significant correlation between miR‐92b expression and DAB2IP expression in GC tissues. Conclusions Our results suggest that miR‐92b promotes GC cell proliferation by activating the DAB2IP‐mediated PI3K/AKT signalling pathway. The miR‐92b/DAB2IP/PI3K/AKT signalling axis may be a potential therapeutic target to prevent GC progression.
Collapse
Affiliation(s)
- Qing-Feng Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Zhang
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong, China
| | - Jia-Wei Yu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Ru-Heng Hua
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Qu-Hui Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jian-Wei Zhu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
25
|
Li L, Hua L, Fan H, He Y, Xu W, Zhang L, Yang J, Deng F, Zeng F. Interplay of PKD3 with SREBP1 Promotes Cell Growth via Upregulating Lipogenesis in Prostate Cancer Cells. J Cancer 2019; 10:6395-6404. [PMID: 31772672 PMCID: PMC6856730 DOI: 10.7150/jca.31254] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/12/2019] [Indexed: 12/26/2022] Open
Abstract
Protein kinase D (PKD) has been implicated in cancer cell survival, proliferation, migration and angiogenesis. However, it is still unknown whether PKD regulates cell proliferation through lipid metabolism in cancer cells. Here we report a novel function of PKD3, a member of PKD family, in regulating of prostate cancer cell proliferation by modulation of SREBP1-mediated de novo lipogenesis. We show that silencing of PKD3 significantly reduces lipid content and expression of the lipogenic genes encoding FASN and ATP-citrate lyase (ACLY). Moreover, endogenous PKD3 interacts with sterol regulatory element binding protein 1(SREBP1) in DU145 cells. Interestingly, PKD3 silencing decreases not only the level of matured-SREBP1 (68KD) but also the binding of SREBP1 to the promoter of fasn gene. In addition, overexpression of SREBP1 reverses the suppression of cell growth caused by PKD3 depletion. Finally, immune-histochemical staining indicate that PKD3 expression is positively correlated with expression of FASN and SREBP1 in prostate cancers. Taken together, these data suggest that targeting PKD3-mediated de novo lipogenesis may be a potential therapeutic approach to block prostate cancer progression.
Collapse
Affiliation(s)
- Ling Li
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Liang Hua
- Center Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huihui Fan
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yu He
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wanfu Xu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lin Zhang
- Department of Clinical Laboratory, Foshan women and children hospital, Foshan, China
| | - Jie Yang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fangyin Zeng
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Circ-IGF1R has pro-proliferative and anti-apoptotic effects in HCC by activating the PI3K/AKT pathway. Gene 2019; 716:144031. [PMID: 31377314 DOI: 10.1016/j.gene.2019.144031] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022]
Abstract
Circular RNAs (circRNAs), a novel class of widespread and diverse endogenous RNAs, have been identified as critical regulators of various cancers, including hepatocellular carcinoma (HCC). However, the specific roles of circRNAs in HCC are largely unknown. In this study, we identified a novel circRNA, circ-IGF1R, in HCC tumour tissues and cell lines. Circ-IGF1R levels were found to be significantly upregulated in HCC tissues compared with levels in paired peritumoural tissues. The high expression levels of circ-IGF1R in HCC were associated with tumour size. Moreover, knocking down circ-IGF1R with siRNA significantly attenuated cell proliferation and induced cell apoptosis and cell cycle arrest in vitro. Further investigation revealed that PI3K/AKT signalling pathway activation was involved in the oncogenic functions of circ-IGF1R in HCC. Our study suggests that circ-IGF1R may be a potential target for the prevention and treatment of HCC.
Collapse
|
27
|
Nourbakhsh M, Mansoor A, Koro K, Zhang Q, Minoo P. Expression Profiling Reveals Involvement of WNT Pathway in the Malignant Progression of Sessile Serrated Adenomas. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1732-1743. [PMID: 31199922 DOI: 10.1016/j.ajpath.2019.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023]
Abstract
Approximately 15% to 20% of colorectal cancers are developed through the serrated pathway of tumorigenesis, which is associated with BRAF mutation, CpG island methylation phenotype, and MLH1 methylation. However, the detailed process of progression from sessile serrated adenoma (SSA) to dysplasia and carcinoma has not been elucidated. To further characterize mechanisms involved in the dysplastic progression of SSA, we investigated differential expressions of mRNAs between areas with and without dysplasia within the same SSA polyps. Significantly dysregulated genes in paired samples were applied for functional annotation and biological significance. The same lysates from a subset of matched samples were subjected for miRNA expression profiling. Differentially expressed miRNAs were determined, and their targeted mRNAs were compared in parallel to the list of differentially expressed mRNAs from an RNA sequencing study. Fourteen common mRNA targets were identified, which include AXIN2, a known indicator of WNT/β-catenin pathway activation. Together, in this study, different genes, pathways, and biological processes involved in the initiation and progression of dysplasia in the serrated pathway are documented. One of the most significant findings is the involvement of the WNT/β-catenin pathway in the dysplastic progression of SSAs with different genes being targeted in early versus advanced dysplasia.
Collapse
Affiliation(s)
- Mahra Nourbakhsh
- Department of Pathology, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Adnan Mansoor
- Department of Pathology, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Konstantin Koro
- Department of Pathology, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Qingrun Zhang
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Parham Minoo
- Department of Pathology, Cumming School of Medicine and Alberta Public Laboratories, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
28
|
Wang T, Cheng Y, Han H, Liu J, Tian B, Liu X. miR-194 Accelerates Apoptosis of Aβ 1⁻42-Transduced Hippocampal Neurons by Inhibiting Nrn1 and Decreasing PI3K/Akt Signaling Pathway Activity. Genes (Basel) 2019; 10:genes10040313. [PMID: 31010100 PMCID: PMC6523401 DOI: 10.3390/genes10040313] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022] Open
Abstract
This article explores the mechanism of miR-194 on the proliferation and apoptosis of Aβ1–42-transduced hippocampal neurons. Aβ1–42-transduced hippocampal neuron model was established by inducing hippocampal neurons with Aβ1–42. MTT assay and flow cytometry were used to detect the viability and apoptosis of hippocampal neurons, respectively. qRT-PCR was used to detect changes in miR-194 and Nrn1 expression after Aβ1–42 induction. Aβ1–42-transduced hippocampal neurons were transfected with miR-194 mimics and/or Nrn1 overexpression vectors. Their viability and neurite length were detected by MTT assay and immunofluorescence, respectively. Western blot was used to detect protein expression. Aβ1–42 inhibited Aβ1–42-transduced hippocampal neuron activity and promoted their apoptosis in a dose-dependent manner. miR-194 was upregulated and Nrn1 was downregulated in Aβ1–42-transduced hippocampal neurons (p < 0.05). Compared with the model group, Aβ1–42-transduced hippocampal neurons of the miR-194 mimic group had much lower activity, average longest neurite length, Nrn1, p-AkT, and Bcl-2 protein expression and had much higher Bax, Caspase-3, and Cleaved Caspase-3 protein expression. Compared with the model group, Aβ1–42-transduced hippocampal neurons of the LV-Nrn1 group had much higher activity, average longest neurite length, Nrn1, p-AkT, and Bcl-2 protein expression and had much lower Bax, Caspase-3, and Cleaved Caspase-3 protein expression. Nrn1 is a target gene of miR-194. miR-194 inhibited apoptosis of Aβ1–42-transduced hippocampal neurons by inhibiting Nrn1 and decreasing PI3K/AkT signaling pathway activity.
Collapse
Affiliation(s)
- Tingting Wang
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Yaling Cheng
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Haibin Han
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Jie Liu
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Bo Tian
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| | - Xiaocui Liu
- Psychiatric Department V, Qingdao Mental Health Center, No. 299, Nanjing Road, Shibei District, Qingdao 266000, China.
| |
Collapse
|
29
|
Li P, Xiao Z, Luo J, Zhang Y, Lin L. MiR-139-5p, miR-940 and miR-193a-5p inhibit the growth of hepatocellular carcinoma by targeting SPOCK1. J Cell Mol Med 2019; 23:2475-2488. [PMID: 30710422 PMCID: PMC6433657 DOI: 10.1111/jcmm.14121] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/26/2022] Open
Abstract
The study was aimed to screen out miRNAs with differential expression in hepatocellular carcinoma (HCC), and to explore the influence of the expressions of these miRNAs and their target gene on HCC cell proliferation, invasion and apoptosis. MiRNAs with differential expression in HCC were screened out by microarray analysis. The common target gene of these miRNAs (miR‐139‐5p, miR‐940 and miR‐193a‐5p) was screened out by analysing the target genes profile (acquired from Targetscan) of the three miRNAs. Expression levels of miRNAs and SPOCK1 were determined by quantitative real time polymerase chain reaction (qRT‐PCR). The target relationships were verified by dual luciferase reporter gene assay and RNA pull‐down assay. Through 3‐(4,5‐dimethyl‐2‐thiazolyl)‐2,5‐diphenyl‐2‐H‐tetrazolium bromide,thiazolyl blue tetrazolium bromide (MTT) and transwell assays and flow cytometry, HCC cell viability, invasion and apoptosis were determined. In vivo experiment was conducted in nude mice to investigate the influence of three miRNAs on tumour growth. Down‐regulation of miR‐139‐5p, miR‐940 and miR‐193a‐5p was found in HCC. Overexpression of these miRNAs suppressed HCC cell viability and invasion, promoted apoptosis and inhibited tumour growth. SPOCK1, the common target gene of miR‐139‐5p, miR‐940 and miR‐193a‐5p, was overexpressed in HCC. SPOCK1 overexpression promoted proliferation and invasion, and restrained apoptosis of HCC cells. MiR‐139‐5p, miR‐940 and miR‐193a‐5p inhibited HCC development through targeting SPOCK1.
Collapse
Affiliation(s)
- Peng Li
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhiwei Xiao
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiajun Luo
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yaojun Zhang
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Lizhu Lin
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Zhang Y, Chen P, Yin W, Ji Y, Shen Q, Ni Q. Nectin-4 promotes gastric cancer progression via the PI3K/AKT signaling pathway. Hum Pathol 2018; 72:107-116. [PMID: 29208564 DOI: 10.1016/j.humpath.2017.10.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/09/2017] [Accepted: 10/18/2017] [Indexed: 02/07/2023]
Abstract
Nectin-4, a member of the Nectin family that includes 4 Ca+-independent immunoglobulin-like cell adhesion molecules, plays a carcinogenic role in multiple cancers. However, Nectin-4 expression and its biological role in gastric cancer (GC) remain largely unknown. In this study, quantitative real-time polymerase chain reaction, Western blotting, and immunohistochemistry were used to evaluate the expression patterns of Nectin-4 in GC specimens and cell lines. We observed that high expression of Nectin-4 in GC patients was associated with TNM stage and lymph node metastasis status, and poor prognosis. In addition, cell proliferation and cell migration assays in vitro and tumorigenicity in vivo were performed to observe the effects of up-regulation and down-regulation of Nectin-4 expression on GC cell phenotypes. In further studies, the PI3K/AKT signaling pathway was revealed to be involved in Nectin-4-mediated GC progression. These results demonstrated that Nectin-4 had a promoter effect on human GC cell growth and motility, indicating that Nectin-4 may serve as an effective therapeutic target in GC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Peisheng Chen
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Wei Yin
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Ye Ji
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Qin Shen
- Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Qingfeng Ni
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
31
|
Koshizuka K, Hanazawa T, Kikkawa N, Katada K, Okato A, Arai T, Idichi T, Osako Y, Okamoto Y, Seki N. Antitumor miR-150-5p and miR-150-3p inhibit cancer cell aggressiveness by targeting SPOCK1 in head and neck squamous cell carcinoma. Auris Nasus Larynx 2017; 45:854-865. [PMID: 29233721 DOI: 10.1016/j.anl.2017.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/04/2017] [Accepted: 11/29/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Our recent studies have revealed that both strands of pre-miRNAs, the guide strand and the passenger strand, are involved in cancer pathogenesis. Analyses of miRNA expression signatures by RNA sequencing in head and neck squamous cell carcinoma (HNSCC) showed that both of the strands of pre-miR-150 (miR-150-5p and miR-150-3p) were significantly downregulated, and that these miRNAs acted as antitumor miRNAs in HNSCC cells. The aim of this study was to identify oncogenic genes in HNSCC cells that were regulated by miR-150-5p and miR-150-3p. METHODS Genome-wide gene expression studies, in silico analyses and dual-luciferase reporter assays were carried out to predict miR-150-5p and miR-150-3p regulation in HNSCC cells. Knockdown assay was applied to investigate the functional significance of the target gene. Overall patient survival as a function of target gene expression was estimated by The Cancer Genome Atlas (TCGA) database. RESULTS A total of 19 genes were putative targets of both miR-150-5p and miR-150-3p regulation. Among them, SPOCK1 (SPARC/osteonectin, cwcv and kazal-like domains proteoglycan 1) was directly regulated by both miRNAs in HNSCC cells. Knockdown studies using si-SPOCK1 showed that expression of SPOCK1 enhanced HNSCC cell aggressiveness. Overexpression of SPOCK1/SPOCK1 was confirmed in HNSCC clinical specimens. Interestingly, analysis of a large number of patients in the TCGA database (n=248) demonstrated that patients with high SPOCK1 expression had significantly shorter survival than did those with low SPOCK1 expression (P=0.0003). Moreover, 15 pathways were identified as SPOCK1-mediated downstream pathways. CONCLUSION Downregulation of both strands of pre-miR-150 (miR-150-5p and miR-150-3p) and overexpression of SPOCK1 contribute to the aggressive nature of HNSCC. The involvement of passenger strand miRNA in the regulation of HNSCC pathogenesis is a novel concept in RNA research.
Collapse
Affiliation(s)
- Keiichi Koshizuka
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan; Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naoko Kikkawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Koji Katada
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Okato
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takayuki Arai
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tetsuya Idichi
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Yusaku Osako
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima, Japan
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Naohiko Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
32
|
Theocharis AD, Karamanos NK. Proteoglycans remodeling in cancer: Underlying molecular mechanisms. Matrix Biol 2017; 75-76:220-259. [PMID: 29128506 DOI: 10.1016/j.matbio.2017.10.008] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023]
Abstract
Extracellular matrix is a highly dynamic macromolecular network. Proteoglycans are major components of extracellular matrix playing key roles in its structural organization and cell signaling contributing to the control of numerous normal and pathological processes. As multifunctional molecules, proteoglycans participate in various cell functions during morphogenesis, wound healing, inflammation and tumorigenesis. Their interactions with matrix effectors, cell surface receptors and enzymes enable them with unique properties. In malignancy, extensive remodeling of tumor stroma is associated with marked alterations in proteoglycans' expression and structural variability. Proteoglycans exert diverse functions in tumor stroma in a cell-specific and context-specific manner and they mainly contribute to the formation of a permissive provisional matrix for tumor growth affecting tissue organization, cell-cell and cell-matrix interactions and tumor cell signaling. Proteoglycans also modulate cancer cell phenotype and properties, the development of drug resistance and tumor stroma angiogenesis. This review summarizes the proteoglycans remodeling and their novel biological roles in malignancies with particular emphasis to the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26500 Patras, Greece.
| |
Collapse
|
33
|
Gao L, Qiu H, Liu J, Ma Y, Feng J, Qian L, Zhang J, Liu Y, Bian T. KLF15 promotes the proliferation and metastasis of lung adenocarcinoma cells and has potential as a cancer prognostic marker. Oncotarget 2017; 8:109952-109961. [PMID: 29299121 PMCID: PMC5746356 DOI: 10.18632/oncotarget.21972] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 09/24/2017] [Indexed: 12/19/2022] Open
Abstract
Lung adenocarcinoma (LADC)is a general form of non-small cell lung cancer that represents a significant threat to public health worldwide. The 5-year-survival rate for LADC is currently below 15%. The transcription factor KLF15, also called kidney-enriched KLF (KKLF), has been proven to play a role in inhibiting proliferation and diversification of carcinoma cells, including those of the endometrium, pancreas and breast, but the involvement of KLF15 in LADC has not previously been studied. In this study, we compared the in vitro expression of KLF15 in human LADC tissues and adjacent normal lung tissues. Expression of KLF15 was found to be abnormally high in LADC tissues and cells compared with adjacent non-tumorous tissues, and was correlated with tumor TNM stage and tumor differentiation (P = 0.003, P = 0.001, respectively). The effect of KLF15 on cell growth and migration were explored in vitro by Western Blotting, CCK8 and colony formation assays, flow cytometry analysis and transwell migration assays, and in vivo by analysis of tumorigenesis in 5-week old BALB/c nude mice. Knockdown of KLF15 significantly upregulated the protein levels of cleaved caspase-3, caspase-7, caspase-8 and PARP, thereby inducing apoptosis. Downregulation of KLF15 in A549 and NCI-H1650 cell lines resulted in these cell lines exhibiting markedly slower growth rates when injected subcutaneously into the flank of nude mice, compared with the comparator control groups (P < 0.05). Collectively, our findings suggest that KLF15 may have a significant effect on LADC cell survival, and that it represents a potential therapeutic and preventive biomarker for LADC prognosis and treatment.
Collapse
Affiliation(s)
- Lihua Gao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Hongmei Qiu
- Department of Respiration, Nantong Geriatric Rehabilitation Hospital, Branch of Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jian Liu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yuzhen Ma
- Centre of Reproductive Medicine, Inner Mongolia Hospital, Inner Mongolia, Hohhot, 010021, China
| | - Jia Feng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Li Qian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
34
|
Chen D, Zhou H, Liu G, Zhao Y, Cao G, Liu Q. SPOCK1 promotes the invasion and metastasis of gastric cancer through Slug-induced epithelial-mesenchymal transition. J Cell Mol Med 2017; 22:797-807. [PMID: 28940639 PMCID: PMC5783867 DOI: 10.1111/jcmm.13357] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/20/2017] [Indexed: 01/05/2023] Open
Abstract
Metastasis is a crucial impediment to the successful treatment for gastric cancer. SPOCK1 has been demonstrated to facilitate cancer metastasis in certain types of cancers; however, the role of SPOCK1 in the invasion and metastasis of gastric cancer remains elusive. SPOCK1 and epithelial-mesenchymal transition (EMT)-related biomarkers were detected by immunohistochemistry and Western blot in gastric cancer specimens. Other methods including stably transfected against SPOCK1 into gastric cancer cells, Western blot, migration and invasion assays in vitro and metastasis assay in vivo were also performed. The elevated expression of SPOCK1 correlates with EMT-related markers in human gastric cancer tissue, clinical metastasis and a poor prognosis in patients with gastric cancer. In addition, knockdown of SPOCK1 expression significantly inhibits the invasion and metastasis of gastric cancer cells in vitro and in vivo, inversely, SPOCK1 overexpression results in the opposite effect. Interestingly, SPOCK1 expression has no effect on cell proliferation in vitro and in vivo. Regarding the mechanism(s) of SPOCK1-induced cells invasion and metastasis, we prove that Slug-induced EMT is involved in SPOCK1-facilitating gastric cancer cells invasion and metastasis. The elevated SPOCK1 expression is closely correlated with cancer metastasis and patient survival, and SPOCK1 promotes the invasion and metastasis of gastric cancer through Slug-mediated EMT, thereby possibly providing a novel therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Dehu Chen
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, Jiangsu, China
| | - Haihua Zhou
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, Jiangsu, China
| | - Guiyuan Liu
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, Jiangsu, China
| | - Yinghai Zhao
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, Jiangsu, China
| | - Gan Cao
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, Jiangsu, China
| | - Qinghong Liu
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, Jiangsu, China
| |
Collapse
|
35
|
Wang Y, Wang W, Qiu E. SPOCK1 promotes the growth of Osteosarcoma cells through mTOR-S6K signaling pathway. Biomed Pharmacother 2017; 95:564-570. [PMID: 28869894 DOI: 10.1016/j.biopha.2017.08.116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/20/2017] [Accepted: 08/24/2017] [Indexed: 12/17/2022] Open
Abstract
SPOCK1 belongs to the SPARC family, which plays an important role in proliferation, invasion and migration of various tumour cells. However, the functions of SPOCK1 in osteosarcoma cell growth and proliferation have not been fully elucidated. In the present study, we found that SPOCK1 is significantly upregulated in osteosarcoma tissue. Moreover, overexpression of SPOCK1 was associated with tumour size, metastasis, Enneking stage and pathological degree. Furthermore, knockdown of SPOCK1 expression suppressed the growth of osteosarcoma cells in vitro and reduced tumourigenicity in nude mice in vivo. Additionally, our data suggest that inactivation of the mTOR-S6K signaling pathway participated in inhibition of SPOCK1-mediated suppression of osteosarcoma cell growth. These findings represent a novel pathogenetic mechanism of osteosarcoma development that provides a potential target for therapeutic strategies for osteosarcoma.
Collapse
Affiliation(s)
- Yuming Wang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, China.
| | - Wei Wang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, China
| | - Enduo Qiu
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, China
| |
Collapse
|