1
|
Sangthong J, Thuwajit C, Jensen LT, Komyod W, Yuvaniyama J, Ponglikitmongkol M. BRCA1 deficiency enhances the aggressiveness of breast cancer cells expressing HPV16 oncoproteins. Biol Cell 2024; 116:e202300072. [PMID: 38514439 DOI: 10.1111/boc.202300072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND INFORMATION The precise etiology of breast cancer is not completely understood, although women with BRCA1 gene mutations have a significantly increased risk of developing the disease. In addition, sporadic breast cancer is frequently associated with decreased BRCA1 gene expression. Growing evidence of Human papillomaviruses (HPVs) infections in breast tumors has raised the possibility of the involvement of HPVs in the pathogenesis of breast cancer. We investigated whether the effects of HPV oncoproteins E6 and E7 were influenced by the expression levels of BRCA1. HPV16E6E7 (prototype or E6D25E/E7N29S Asian variant type) were stably expressed in MDA-MB231 breast cancer cells, wild type for BRCA1, or with BRCA1 knocked down. RESULTS Expression of HPV16E6E7 oncogenes did not affect BRCA1 levels and the abundance of HPV16E6E7 was not altered by BRCA1 knockdown. BRCA1 levels did not alter HPV16E6E7-dependent degradation of G1-S cell cycle proteins p53 and pRb. However, we found that the expression of G2-M cell cycle protein cyclin B1 enhanced by HPV16E6E7 was impacted by BRCA1 levels. Especially, we found the correlation between BRCA1 and cyclin B1 expression and this was also confirmed in breast cancer samples from a Thai cohort. We further demonstrated that the combination of HPV oncoproteins and low levels of BRCA1 protein appears to enhance proliferation and invasion. Transactivation activities of HPV16E6E7 on genes regulating cell proliferation and invasion (TGF-β and vimentin) were significantly increased in BRCA1-deficient cells. CONCLUSIONS Our results indicate that a deficiency of BRCA1 promotes the transactivation activity of HPV16E6E7 leading to increase of cell proliferation and invasion. SIGNIFICANCE HPV infection appears to have the potential to enhance the aggressiveness of breast cancers, especially those deficient in BRCA1.
Collapse
Affiliation(s)
- Jariya Sangthong
- Faculty of Science, Graduate Program in Molecular Medicine, Mahidol University, Bangkok, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand
| | - Laran T Jensen
- Faculty of Science, Department of Biochemistry, Mahidol University, Bangkok, Thailand
| | - Waraporn Komyod
- Faculty of Science, Department of Biochemistry, Mahidol University, Bangkok, Thailand
| | - Jirundon Yuvaniyama
- Faculty of Science, Department of Biochemistry, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
2
|
Cavalcante IL, Silva Barros CCD, Colares DF, Cruz VMS, de Andrade BAB, Nonaka CFW, Rabenhorst SHB, Cavalcante RB. BubR1 and cyclin B1 immunoexpression in pleomorphic adenoma and polymorphous adenocarcinoma of minor salivary glands. Pathol Res Pract 2024; 253:154961. [PMID: 38043194 DOI: 10.1016/j.prp.2023.154961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023]
Abstract
The immunoexpression of BubR1 and cyclin B1 in pleomorphic adenoma (PA) and polymorphic adenocarcinoma (PAC) in minor salivary glands is poorly studied. Thus, a retrospective and observational study was performed to provide a better understanding of the role and immunopositivity patterns of these proteins in these lesions. Sixteen cases of PA and 16 cases of PAC were selected. Parenchyma cells were submitted to quantitative immunohistochemical analysis through the labeling index. Cytoplasmic immunoexpression of BubR1 was observed in neoplastic cells from all analyzed PA and PAC cases. All PA cases and 93.7% of PAC exhibited nuclear immunoexpression of BubR1. Higher cytoplasmic and nuclear immunoexpression of BubR1 was observed in PAC (p = 0.001 and p = 0.122, respectively). Cytoplasmic immunoexpression of cyclin B1 was observed in all cases of PA and PAC, with a higher labeling index in the latter (p < 0.001). There was a significant positive correlation between nuclear and cytoplasmic BubR1 immunoexpressions (p < 0.001) in PA and a significant negative correlation between BubR1 and cyclin B1 cytoplasmic immunoexpressions (p = 0.014) in PAC. The higher cytoplasmic and nuclear immunoexpression of BubR1 in PACs suggests the continuous maintenance of neoplastic cells in the cell cycle and migration. Higher immunoexpression of cyclin B1 supports this lesion's enhanced proliferative and migration ability.
Collapse
Affiliation(s)
- Israel Leal Cavalcante
- Oral Pathology Section, Department of Dentistry, University of Fortaleza, Fortaleza, CE, Brazil; Department of Oral Diagnosis and Pathology, School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Caio César da Silva Barros
- Postgraduate Program in Dental Sciences, Oral Pathology and Medicine, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Débora Frota Colares
- Postgraduate Program in Dental Sciences, Oral Pathology and Medicine, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Vitória Maria Sousa Cruz
- Oral Pathology Section, Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | | | | | - Silvia Helena Barem Rabenhorst
- Postgraduate Program in Biological Sciences, Molecular Genetics Laboratory, Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | |
Collapse
|
3
|
Xiao B, Xiang Q, Deng Z, Chen D, Wu S, Zhang Y, Liang Y, Wei S, Luo G, Li L. KCNN1 promotes proliferation and metastasis of breast cancer via ERLIN2-mediated stabilization and K63-dependent ubiquitination of Cyclin B1. Carcinogenesis 2023; 44:809-823. [PMID: 37831636 PMCID: PMC10818095 DOI: 10.1093/carcin/bgad070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Potassium Calcium-Activated Channel Subfamily N1 (KCNN1), an integral membrane protein, is thought to regulate neuronal excitability by contributing to the slow component of synaptic after hyperpolarization. However, the role of KCNN1 in tumorigenesis has been rarely reported, and the underlying molecular mechanism remains unclear. Here, we report that KCNN1 functions as an oncogene in promoting breast cancer cell proliferation and metastasis. KCNN1 was overexpressed in breast cancer tissues and cells. The pro-proliferative and pro-metastatic effects of KCNN1 were demonstrated by CCK8, clone formation, Edu assay, wound healing assay and transwell experiments. Transcriptomic analysis using KCNN1 overexpressing cells revealed that KCNN1 could regulate key signaling pathways affecting the survival of breast cancer cells. KCNN1 interacts with ERLIN2 and enhances the effect of ERLIN2 on Cyclin B1 stability. Overexpression of KCNN1 promoted the protein expression of Cyclin B1, enhanced its stability and promoted its K63 dependent ubiquitination, while knockdown of KCNN1 had the opposite effects on Cyclin B1. Knockdown (or overexpression) ERLNI2 partially restored Cyclin B1 stability and K63 dependent ubiquitination induced by overexpression (or knockdown) of KCNN1. Knockdown (or overexpression) ERLIN2 also partially neutralizes the effects of overexpression (or knockdown) KCNN1-induced breast cancer cell proliferation, migration and invasion. In paired breast cancer clinical samples, we found a positive expression correlations between KCNN1 and ERLIN2, KCNN1 and Cyclin B1, as well as ERLIN2 and Cyclin B1. In conclusion, this study reveals, for the first time, the role of KCNN1 in tumorigenesis and emphasizes the importance of KCNN1/ERLIN2/Cyclin B1 axis in the development and metastasis of breast cancer.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| | - Zihua Deng
- Department of General Surgery Section 5, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511518, China
| | - Daxiang Chen
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| | - Yanxia Zhang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| | - Yaru Liang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| | - Shi Wei
- Department of Histology and Embryology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guoqing Luo
- Department of General Surgery Section 5, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan 511518, China
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong 511518, China
| |
Collapse
|
4
|
O’Leary BR, Kalen AL, Pope AN, Goswami PC, Cullen JJ. Hydrogen Peroxide Mediates Pharmacological Ascorbate Induced Radio-Sensitization of Pancreatic Cancer Cells by Enhancing G2-accumulation and Reducing Cyclin B1 Protein Levels. Radiat Res 2023; 200:444-455. [PMID: 37758045 PMCID: PMC10699322 DOI: 10.1667/rade-22-00182.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 08/24/2023] [Indexed: 10/03/2023]
Abstract
Pharmacological ascorbate (P-AscH-, high dose, intravenous vitamin C) preferentially sensitizes human pancreas ductal adenocarcinoma (PDAC) cells to radiation-induced toxicity compared to non-tumorigenic epithelial cells. Radiation-induced G2-checkpoint activation contributes to the resistance of cancer cells to DNA damage induced toxicity. We hypothesized that P-AscH- induced radio-sensitization of PDAC cells is mediated by perturbations in the radiation induced activation of the G2-checkpoint pathway. Both non-tumorigenic pancreatic ductal epithelial and PDAC cells display decreased clonogenic survival and increased doubling times after radiation treatment. In contrast, the addition of P-AscH- to radiation increases clonogenic survival and decreases the doubling time of non-tumorigenic epithelial cells but decreasing clonogenic survival and increasing the doubling time of PDAC cells. Results from the mitotic index and propidium iodide assays showed that while the P-AscH- treatments did not affect radiation-induced G2-checkpoint activation, it enhanced G2-accumulation. The addition of catalase reverses the increases in G2-accumulation, indicating a peroxide-mediated mechanism. In addition, P-AscH- treatment of PDAC cells suppresses radiation-induced accumulation of cyclin B1 protein levels. Both translational and post-translational pathways appear to regulate cyclin B1 protein levels after the combination treatment of PDAC cells with P-AscH- and radiation. The protein changes seen are reversed by the addition of catalase suggesting that hydrogen peroxide mediates P-AscH- induced radiation sensitization of PDAC cells by enhancing G2-accumulation and reducing cyclin B1 protein levels.
Collapse
Affiliation(s)
- Brianne R. O’Leary
- Departments of Surgery and Free Radical and Radiation Biology Division, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Amanda L. Kalen
- Department of Radiation Oncology, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Amanda N. Pope
- Department of Radiation Oncology, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Prabhat C. Goswami
- Department of Radiation Oncology, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Joseph J. Cullen
- Departments of Surgery and Free Radical and Radiation Biology Division, The University of Iowa Carver College of Medicine, Iowa City, Iowa
- Department of Radiation Oncology, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
5
|
Wang XX, Wu HY, Yang Y, Ma MM, Zhang YW, Huang HZ, Li SH, Pan SL, Tang J, Peng JH. CCNB1 is involved in bladder cancer pathogenesis and silencing CCNB1 decelerates tumor growth and improves prognosis of bladder cancer. Exp Ther Med 2023; 26:382. [PMID: 37456156 PMCID: PMC10347295 DOI: 10.3892/etm.2023.12081] [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: 02/14/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
In search of an effective therapeutic target for bladder urothelial carcinoma (BLCA), the present study aimed to investigate the expression of cyclin B1 (CCNB1) and its putative mechanism in BLCA. BLCA sequencing data from Gene Expression Omnibus and The Cancer Genome Atlas were used to analyze expression of CCNB1 mRNA and high CCNB1 expression had a poorer prognosis compared with those with low expression. Immunohistochemistry (IHC) samples collected from the Human Protein Atlas database were analyzed for CCNB1 protein expression. Short hairpin (sh) CCNB1-transfected BLCA T24 and 5637 cells were used to investigate the effects of CCNB1 and inhibit the proliferation, migration and invasion of BLCA cells, affect the cell cycle distribution and promote apoptosis of 5637 cells. A sh-CCNB1 BLCA chicken embryo chorioallantoic membrane (CAM) transplantation model was established to observe the impacts of sh-CCNB1 on the tumorigenesis of BLCA in vivo. Analysis of sequencing data showed that CCNB1 mRNA was significantly elevated in tumor and BLCA compared with normal tissues [standardized mean difference (SMD)=1.21; 95% CI: 0.26-2.15; I²=95.9%]. IHC indicated that CCNB1 protein was localized in the nucleus and cytoplasm and was significantly increased in BLCA tumor tissues. The in vitro tests demonstrated that proliferation of T24 and 5637 cells transfected with sh-CCNB1 was significantly inhibited and cell migration and invasion ability were significantly decreased. sh-CCNB1 decreased the percentage of T24 cells in G0/G1, 5637 cells in the G0/G1 phase and S phase and increased percentage of 5637 cells in the G2/M phase and increased early apoptosis of 5637 cells. The in vivo experiments demonstrated that the mass of transplanted tumors was significantly decreased compared with the control group following silencing of CCNB1. The present results suggested that CCNB1 was involve in the development and prognosis of BLCA and silencing of CCNB1 may be a promising targeted therapy for BLCA.
Collapse
Affiliation(s)
- Xue-Xuan Wang
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Key Laboratory of Longevity and Aging-Related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Hua-Yu Wu
- Medical Experimental Center, The First People's Hospital of Nanning, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, P.R. China
| | - Ying Yang
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Miao-Miao Ma
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yi-Wei Zhang
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hai-Zhen Huang
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Sheng-Hua Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shang-Ling Pan
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Key Laboratory of Longevity and Aging-Related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Jun Tang
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Key Laboratory of Longevity and Aging-Related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| | - Jun-Hua Peng
- Department of Pathophysiology, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Key Laboratory of Longevity and Aging-Related Diseases, Guangxi Medical University, Ministry of Education, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
6
|
Aljohani AI, Toss MS, Green AR, Rakha EA. The clinical significance of cyclin B1 (CCNB1) in invasive breast cancer with emphasis on its contribution to lymphovascular invasion development. Breast Cancer Res Treat 2023; 198:423-435. [PMID: 36418517 PMCID: PMC10036284 DOI: 10.1007/s10549-022-06801-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Lymphovascular invasion (LVI) is regulated through complex molecular mechanisms. Cyclin B1 (CCNB1) was previously determined as being associated with LVI using large cohorts of breast cancer (BC) and artificial neural network (ANN) technique. In this study, we aimed to assess the association between CCNB1 and LVI, other clinicopathological and other LVI-related biomarkers at the molecular (RNA transcriptomic) and proteomic levels in BC. METHODS Two transcriptomic BC cohorts (n = 2834) were used to assess the association between the expression of CCNB1 at the mRNA level and clinicopathological characteristics and patient outcome. Tissue microarrays (TMAs) from a well-characterised BC cohort (n = 2480) with long-term outcome were also used to assess the clinical significance of CCNB1 protein expression using immunohistochemistry. RESULTS High CCNB1 mRNA expression was associated with aggressive tumour behaviour, including LVI, larger size, higher tumour grade, high lymph nodal stage, hormonal receptor negativity, HER2 positivity and poor clinical outcome (all p < 0.0001). Similarly, high CCNB1 protein expression was associated with higher tumour grade, hormonal receptor negativity and HER2 positivity (all p < 0.0001). Additionally, there was a significant association between CCNB1- and LVI-related biomarkers including N-cadherin, P-cadherin and TWIST2 at the transcriptomic and proteomic level. Multivariate analysis revealed that CCNB1 was an independent predictor of shorter BC-specific survival (HR = 1.3; 95% CI 1.2-1.5; p = 0.010). CONCLUSION CCNB1 is a key gene associated with LVI in BC and has prognostic value. More functional studies are warranted to unravel the mechanistic role of CCNB1 in the development of LVI.
Collapse
Affiliation(s)
- Abrar I Aljohani
- Academic Unit for Translational Medical Sciences, School of Medicine, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Michael S Toss
- Academic Unit for Translational Medical Sciences, School of Medicine, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Andrew R Green
- Academic Unit for Translational Medical Sciences, School of Medicine, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK
| | - Emad A Rakha
- Academic Unit for Translational Medical Sciences, School of Medicine, Nottingham Breast Cancer Research Centre, University of Nottingham Biodiscovery Institute, University Park, Nottingham, NG7 2RD, UK.
- Histopathology Department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt.
- Department of Histopathology, Nottingham University Hospital NHS Trust, City Hospital Campus, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
7
|
Nair NU, Jiang Q, Wei JS, Misra VA, Morrow B, Kesserwan C, Hermida LC, Lee JS, Mian I, Zhang J, Lebensohn A, Miettinen M, Sengupta M, Khan J, Ruppin E, Hassan R. Genomic and transcriptomic analyses identify a prognostic gene signature and predict response to therapy in pleural and peritoneal mesothelioma. Cell Rep Med 2023; 4:100938. [PMID: 36773602 PMCID: PMC9975319 DOI: 10.1016/j.xcrm.2023.100938] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/23/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023]
Abstract
Malignant mesothelioma is an aggressive cancer with limited treatment options and poor prognosis. A better understanding of mesothelioma genomics and transcriptomics could advance therapies. Here, we present a mesothelioma cohort of 122 patients along with their germline and tumor whole-exome and tumor RNA sequencing data as well as phenotypic and drug response information. We identify a 48-gene prognostic signature that is highly predictive of mesothelioma patient survival, including CCNB1, the expression of which is highly predictive of patient survival on its own. In addition, we analyze the transcriptomics data to study the tumor immune microenvironment and identify synthetic-lethality-based signatures predictive of response to therapy. This germline and somatic whole-exome sequencing as well as transcriptomics data from the same patient are a valuable resource to address important biological questions, including prognostic biomarkers and determinants of treatment response in mesothelioma.
Collapse
Affiliation(s)
- Nishanth Ulhas Nair
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Qun Jiang
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | | | | | - Betsy Morrow
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | | | - Leandro C Hermida
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joo Sang Lee
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA; School of Medicine and Department of Artificial Intelligence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Idrees Mian
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Jingli Zhang
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | | | | | - Manjistha Sengupta
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Javed Khan
- Genetics Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, CCR, NCI, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
8
|
Recombinant human p53 adenovirus injection combined with Bortezomib inhibits proliferation and promotes apoptosis in multiple myeloma. Leuk Res 2023; 127:107041. [PMID: 36801701 DOI: 10.1016/j.leukres.2023.107041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Multiple myeloma (MM) is a B-cell malignancy characterized by abnormal proliferation of clonal plasma cells in the bone marrow, the incidence of which has further increased in recent years. In multiple myeloma, wild-type functional p53 is often inactivated or dysregulated. Therefore, this study aimed to investigate the role of p53 knockdown or overexpression in multiple myeloma and the therapeutic effect of recombinant adenovirus-p53 (rAd-p53) in combination with Bortezomib. METHODS SiRNA p53 and rAd-p53 were used to knock down and overexpress p53. RT-qPCR was used to detect gene expression, and western blotting (WB) was used to detect protein expression levels. We also constructed wild-type multiple myeloma cell line-MM1S cell xenograft tumor models and explored the effects of siRNA-p53, rAd-p53, and Bortezomib on multiple myeloma in vivo and in vitro. H&E staining and KI67 immunohistochemical staining were used to assess the anti-myeloma effects of recombinant adenovirus and Bortezomib in vivo. RESULTS The designed siRNA p53 effectively led to the knockdown of the p53 gene, while rAd-p53 could significantly achieve p53 overexpression. p53 gene inhibited MM1S cell proliferation and promoted apoptosis of wild-type multiple myeloma cell line MM1S. P53 gene inhibited tumor proliferation in vitro by promoting p21 expression and reducing cell cycle protein B1 expression of MM1S. P53 gene overexpression could inhibit tumor growth in vivo. Injection of rAd-p53 in tumor models inhibited tumor development through p21- and cyclin B1-mediated cell proliferation and apoptosis regulation. CONCLUSIONS We found that overexpression of p53 inhibits MM tumor cell survival and proliferation in vivo and in vitro. Furthermore, the combination of rAd-p53 and Bortezomib significantly improved the efficacy, which provides a new possibility for more effective treatment of MM.
Collapse
|
9
|
Kim YW, Bak SB, Baek SY, Kim IK, Lee WY, Yun UJ, Park KI. Mylabris phalerata induces the apoptosis and cell cycle delay in HCC, and potentiates the effect of sorafenib based on the molecular and network pharmacology approach. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00300-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
10
|
Fu H, Li K, Wang S, Li Y. High expression of CCNB1 driven by ncRNAs is associated with a poor prognosis and tumor immune infiltration in breast cancer. Aging (Albany NY) 2022; 14:6780-6795. [PMID: 36040381 PMCID: PMC9467392 DOI: 10.18632/aging.204253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUNDS Breast cancer (BC) is the most frequent cancer diagnosed in women throughout the world. The purpose of this study was to explore new biomarkers for breast cancer diagnosis. CyclinB1 (CCNB1) is found in abundance in a wide range of human malignancies. MATERIAL AND METHODS We evaluated the transcriptional, survival data and expression levels in tissue data of CCNB1 in patients with breast cancer from the Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), The Human Protein Atlas (THAP) and Genome Tissue Expression (GTEx) database. A series of in silico analyses were used to investigate at noncoding RNAs (ncRNAs), gene ontology (GO) annotation analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG), and the Protein-Protein Interaction (PPI) network. A quantitative real-time polymerase chain reaction (qRT-PCR) was used to validate CCNB1 in BC cell lines. RESULTS CCNB1 expression was higher in BC tissues than in normal breast tissues. It was significantly related to survival time, tumor mutation burden (TMB), methylated, immune cell infiltration, and the expressed in estrogen receptor (ER) (-), lymphnode (+), and p53 (+) groups in BC. Moreover, The AC026401.3/CCNB1-miR-139-5p axis was discovered as the most promising upstream ncRNA-related pathway of CCNB1 in BC. CONCLUSION CCNB1 can be used as an independent predictive factor for BC, indicating that this would be a target for highly precise therapy and a biomarker for the disease.
Collapse
Affiliation(s)
- Hongtao Fu
- Department of Breast Surgery, Jiangsu Province Hospital, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Kun Li
- Department of emergency, Changsha Central Hospital, The Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Shui Wang
- Department of Breast Surgery, Jiangsu Province Hospital, The First Hospital Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yuming Li
- Department of Traditional Chinese Medicine and Western Medicine, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410006, China
| |
Collapse
|
11
|
Fan X, Zhao Z, Ma L, Huang X, Zhan Q, Song Y. PTBP1 promotes IRES-mediated translation of cyclin B1 in cancer. Acta Biochim Biophys Sin (Shanghai) 2022; 54:696-707. [PMID: 35643957 PMCID: PMC9828304 DOI: 10.3724/abbs.2022046] [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: 10/13/2021] [Accepted: 01/11/2022] [Indexed: 11/25/2022] Open
Abstract
Cyclin B1 is an essential cyclin-dependent protein that involves in the G2/M transition. Multiple studies report that cyclin B1 is upregulated in cancers and promotes cancer progression. However, the mechanism of cyclin B1 upregulation remains unclear. Here we report that the 5'UTR of cyclin B1 mRNA contains an internal ribosome entry site (IRES) by using a bicistronic fluorescent reporter. We show that IRES can initiate the translation of cyclin B1, and the IRES-mediated translation is further activated under cell stress. Interacting trans-acting factors (ITAFs) are required by most IRES to initiate the translation. We find that PTBP1 promotes the IRES-mediated translation of cyclin B1 by binding to the 5'UTR of cyclin B1. On top of that, PTBP1 promotes the malignancy of ESCC cells. Our data suggest that the IRES-mediated translation of cyclin B1 plays an essential role in the cyclin B1 upregulation in cancers.
Collapse
Affiliation(s)
- Xinyi Fan
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021 China
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNY10065USA
| | - Zitong Zhao
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021 China
| | - Liying Ma
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021 China
| | | | - Qimin Zhan
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021 China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Laboratory of Molecular OncologyPeking University Cancer Hospital & InstituteBeijing100142China
| | - Yongmei Song
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100021 China
| |
Collapse
|
12
|
Alhashem Z, Feldner-Busztin D, Revell C, Alvarez-Garcillan Portillo M, Camargo-Sosa K, Richardson J, Rocha M, Gauert A, Corbeaux T, Milanetto M, Argenton F, Tiso N, Kelsh RN, Prince VE, Bentley K, Linker C. Notch controls the cell cycle to define leader versus follower identities during collective cell migration. eLife 2022; 11:e73550. [PMID: 35438077 PMCID: PMC9129880 DOI: 10.7554/elife.73550] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
Coordination of cell proliferation and migration is fundamental for life, and its dysregulation has catastrophic consequences, such as cancer. How cell cycle progression affects migration, and vice versa, remains largely unknown. We address these questions by combining in silico modelling and in vivo experimentation in the zebrafish trunk neural crest (TNC). TNC migrate collectively, forming chains with a leader cell directing the movement of trailing followers. We show that the acquisition of migratory identity is autonomously controlled by Notch signalling in TNC. High Notch activity defines leaders, while low Notch determines followers. Moreover, cell cycle progression is required for TNC migration and is regulated by Notch. Cells with low Notch activity stay longer in G1 and become followers, while leaders with high Notch activity quickly undergo G1/S transition and remain in S-phase longer. In conclusion, TNC migratory identities are defined through the interaction of Notch signalling and cell cycle progression.
Collapse
Affiliation(s)
- Zain Alhashem
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College LondonLondonUnited Kingdom
| | | | - Christopher Revell
- Cellular Adaptive Behaviour Lab, Francis Crick InstituteLondonUnited Kingdom
| | | | - Karen Camargo-Sosa
- Department of Biology & Biochemistry, University of BathBathUnited Kingdom
| | - Joanna Richardson
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College LondonLondonUnited Kingdom
| | - Manuel Rocha
- Committee on Development, Regeneration and Stem Cell Biology, The University of ChicagoChicagoUnited States
| | - Anton Gauert
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College LondonLondonUnited Kingdom
| | - Tatianna Corbeaux
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College LondonLondonUnited Kingdom
| | | | | | - Natascia Tiso
- Department of Biology, University of PadovaPadovaItaly
| | - Robert N Kelsh
- Department of Biology & Biochemistry, University of BathBathUnited Kingdom
| | - Victoria E Prince
- Committee on Development, Regeneration and Stem Cell Biology, The University of ChicagoChicagoUnited States
- Department of Organismal Biology and Anatomy, The University of ChicagoChicagoUnited States
| | - Katie Bentley
- Cellular Adaptive Behaviour Lab, Francis Crick InstituteLondonUnited Kingdom
- Department of Informatics, King's College LondonLondonUnited Kingdom
| | - Claudia Linker
- Randall Centre for Cell and Molecular Biophysics, Guy's Campus, King's College LondonLondonUnited Kingdom
| |
Collapse
|
13
|
La Rocca G, Cavalieri V. Roles of the Core Components of the Mammalian miRISC in Chromatin Biology. Genes (Basel) 2022; 13:414. [PMID: 35327968 PMCID: PMC8954937 DOI: 10.3390/genes13030414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 12/16/2022] Open
Abstract
The Argonaute (AGO) and the Trinucleotide Repeat Containing 6 (TNRC6) family proteins are the core components of the mammalian microRNA-induced silencing complex (miRISC), the machinery that mediates microRNA function in the cytoplasm. The cytoplasmic miRISC-mediated post-transcriptional gene repression has been established as the canonical mechanism through which AGO and TNRC6 proteins operate. However, growing evidence points towards an additional mechanism through which AGO and TNRC6 regulate gene expression in the nucleus. While several mechanisms through which miRISC components function in the nucleus have been described, in this review we aim to summarize the major findings that have shed light on the role of AGO and TNRC6 in mammalian chromatin biology and on the implications these novel mechanisms may have in our understanding of regulating gene expression.
Collapse
Affiliation(s)
- Gaspare La Rocca
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90128 Palermo, Italy
| |
Collapse
|
14
|
Yang X, Tian M, Zhang W, Chai T, Shen Z, Kang M, Lin J. Identification of potential core genes in esophageal carcinoma using bioinformatics analysis. Medicine (Baltimore) 2021; 100:e26428. [PMID: 34232175 PMCID: PMC8270608 DOI: 10.1097/md.0000000000026428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a common human malignancy worldwide. The tumorigenesis mechanism in ESCC is unclear. MATERIALS AND METHODS To explore potential therapeutic targets for ESCC, we analyzed 3 microarray datasets (GSE20347, GSE38129, and GSE67269) derived from the gene expression omnibus (GEO) database. Then, the GEO2R tool was used to screen out differently expressed genes (DEGs) between ESCC and normal tissue. Gene ontology function and kyoto encyclopedia of genes and genomes pathway enrichment analysis were performed using the database for annotation, visualization and integrated discovery to identify the pathways and functional annotation of DEGs. Protein-protein interaction of these DEGs was analyzed based on the search tool for the retrieval of interacting genes database and visualized by Cytoscape software. In addition, we used encyclopedia of RNA interactomes (ENCORI), gene expression profiling interactive analysis (GEPIA), and the human protein atlas to confirm the expression of hub genes in ESCC. Finally, GEPIA was used to evaluate the prognostic value of hub genes expression in ESCC patients and we estimated the associations between hub genes expression and immune cell populations (B Cell, CD8+ T Cell, CD4+ T Cell, Macrophage, Neutrophil, and Dendritic Cell) in esophageal carcinoma (ESCA) using tumor immune estimation resource (TIMER). RESULTS In this study, 707 DEGs (including 385 upregulated genes and 322 downregulated genes) and 6 hub genes (cyclin B1 [CCNB1], cyclin dependent kinase 1 [CDK1], aurora kinase A [AURKA], ubiquitin conjugating enzyme E2C [UBE2C], cyclin A2 [CCNA2], and cell division cycle 20 [CDC20]) were identified. All of the 6 hub genes were highly expressed in ESCC tissues. Among of them, only CCNB1 and CDC20 were associated with stage of ESCC and all of them were not associated with survival time of patients. CONCLUSION DEGs and hub genes were confirmed in our study, providing a thorough, scientific and comprehensive research goals for the pathogenesis of ESCC.
Collapse
Affiliation(s)
| | - Mengyue Tian
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | | | - Tianci Chai
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou
- Department of Anesthesiology, Xinyi People's Hospital, Xuzhou
| | | | | | | |
Collapse
|
15
|
Xing Z, Wang X, Liu J, Zhang M, Feng K, Wang X. Expression and prognostic value of CDK1, CCNA2, and CCNB1 gene clusters in human breast cancer. J Int Med Res 2021; 49:300060520980647. [PMID: 33896262 PMCID: PMC8076779 DOI: 10.1177/0300060520980647] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective Cell cycle-associated proteins play important roles in breast cancer (BRCA), based on evidence from cell lines, preclinical murine models, and human tissue samples. Methods Herein, we used the Onomine, GEPIA, Kaplan–Meier Plotter, and cBioPortal databases to examine transcriptional and survival data pertaining to cyclin-associated gene clusters (CDK1, CCNA2, and CCNB1) in BRCA patients. Results CDK1, CCNA2, and CCNB1 gene expression levels were higher in BRCA compared with control tissue samples and were correlated with more-advanced tumor stage. Kaplan–Meier survival analyses confirmed that elevated CDK1, CCNA2, and CCNB1 expression levels were associated with overall and post-progression survival and recurrence-free probability rates in patients with BRCA. Conclusion The results of this study implied that CDK1, CCNA2, and CCNB1 gene clusters may provide potential therapeutic targets and prognostic biomarkers in patients with BRCA.
Collapse
Affiliation(s)
- Zeyu Xing
- Breast Cancer Department, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Breast Cancer Department, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Liu
- Breast Cancer Department, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Menglu Zhang
- Breast Cancer Department, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Feng
- Breast Cancer Department, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Wang
- Breast Cancer Department, National Cancer Center/National Clinical Research Center for Cancer/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Hou Y, Wang X, Wang J, Sun X, Liu X, Hu H, Fan W, Zhang X, Wu D. Cyclin B1 acts as a tumor microenvironment-related cancer promoter and prognostic biomarker in hepatocellular carcinoma. J Int Med Res 2021; 49:3000605211016265. [PMID: 34044639 PMCID: PMC8168034 DOI: 10.1177/03000605211016265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 04/19/2021] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVES The present study aimed to develop a gene signature based on the ESTIMATE algorithm in hepatocellular carcinoma (HCC) and explore possible cancer promoters. METHODS The ESTIMATE and CIBERSORT algorithms were applied to calculate the immune/stromal scores and the proportion of tumor-infiltrating immune cells (TICs) in a cohort of HCC patients. The differentially expressed genes (DEGs) were screened by Cox proportional hazards regression analysis and protein-protein interaction (PPI) network construction. Cyclin B1 (CCNB1) function was verified using experiments. RESULTS The stromal and immune scores were associated with clinicopathological factors and recurrence-free survival (RFS) in HCC patients. In total, 546 DEGs were up-regulated in low score groups, 127 of which were associated with RFS. CCNB1 was regarded as the most predictive factor closely related to prognosis of HCC and could be a cancer promoter. Gene Set Enrichment Analysis (GSEA) and CIBERSORT analyses indicated that CCNB1 levels influenced HCC tumor microenvironment (TME) immune activity. CONCLUSIONS The ESTIMATE signature can be used as a prognosis tool in HCC. CCNB1 is a tumor promoter and contributes to TME status conversion.
Collapse
Affiliation(s)
- Yangming Hou
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xin Wang
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junwei Wang
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xuemei Sun
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinbo Liu
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Han Hu
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenzhe Fan
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinchen Zhang
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Dequan Wu
- Department of Hepatic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
17
|
Hong Z, Wang Q, Hong C, Liu M, Qiu P, Lin R, Lin X, Chen F, Li Q, Liu L, Wang C, Chen D. Identification of Seven Cell Cycle-Related Genes with Unfavorable Prognosis and Construction of their TF-miRNA-mRNA regulatory network in Breast Cancer. J Cancer 2021; 12:740-753. [PMID: 33403032 PMCID: PMC7778540 DOI: 10.7150/jca.48245] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC), with complex tumorigenesis and progression, remains the most common malignancy in women. We aimed to explore some novel and significant genes with unfavorable prognoses and potential pathways involved in BC initiation and progression via bioinformatics methods. BC tissue-specific microarray datasets of GSE42568, GSE45827 and GSE54002, which included a total of 651 BC tissues and 44 normal breast tissues, were obtained from the Gene Expression Omnibus (GEO) database, and 124 differentially expressed genes (DEGs) were identified between BC tissues and normal breast tissues via R software and an online Venn diagram tool. Database for Annotation, Visualization and Integration Discovery (DAVID) software showed that 65 upregulated DEGs were mainly enriched in the regulation of the cell cycle, and Search Tool for the Retrieval of Interacting Genes (STRING) software identified the 39 closest associated upregulated DEGs in protein-protein interactions (PPIs), which validated the high expression of genes in BC tissues by the Gene Expression Profiling Interactive Analysis (GEPIA) tool. In addition, 36 out of 39 BC patients showed significantly worse outcomes by Kaplan-Meier plotter (KM plotter), and an additional Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that seven genes (cyclin E2 (CCNE2), cyclin B1 (CCNB1), cyclin B2 (CCNB2), mitotic checkpoint serine/threonine kinase B (BUB1B), dual-specificity protein kinase (TTK), cell division cycle 20 (CDC20), and pituitary tumor transforming gene 1 (PTTG1)) were markedly enriched in the cell cycle pathway. Analysis of the clinicopathological characteristics of hub genes revealed that seven cell cycle-related genes (CCRGs) were significantly highly expressed in four BC subtypes (luminal A, luminal B, HER2-positive and triple-negative (TNBC)), and except for the CCNE2 gene, high expression levels were significantly associated with tumor pathological grade and stage and metastatic events of BC. Furthermore, genetic mutation analysis indicated that genetic alterations of CCRGs could also significantly affect BC patients' prognosis. A quantitative real-time polymerase chain reaction (qRT-PCR) assay found that the seven CCRGs were significantly differentially expressed in BC cell lines. Integration of published multilevel expression data and a bioinformatics computational approach were used to predict and construct a regulation mechanism: a transcription factor (TF)-microRNA (miRNA)-messenger RNA (mRNA) regulation network. The present work is the first to construct a regulatory network of TF-miRNA-mRNA in BC for CCRGs and provides new insights into the molecular mechanism of BC.
Collapse
Affiliation(s)
- Zhipeng Hong
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China.,Department of Breast Surgery and General Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P. R. China.,Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Qinglan Wang
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Chengye Hong
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Meimei Liu
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Pengqin Qiu
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Rongrong Lin
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Xiaolan Lin
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Fangfang Chen
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Qiuhuang Li
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Lingling Liu
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| | - Chuan Wang
- Department of Breast Surgery and General Surgery, The Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian Province, 350001, P. R. China.,Breast Cancer Institute, Fujian Medical University, Fuzhou, Fujian Province, 350001, P.R. China
| | - Debo Chen
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, P. R. China
| |
Collapse
|
18
|
Ramos J, Yoo C, Felty Q, Gong Z, Liuzzi JP, Poppiti R, Thakur IS, Goel R, Vaid AK, Komotar RJ, Ehtesham NZ, Hasnain SE, Roy D. Sensitivity to differential NRF1 gene signatures contributes to breast cancer disparities. J Cancer Res Clin Oncol 2020; 146:2777-2815. [PMID: 32705365 DOI: 10.1007/s00432-020-03320-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/09/2020] [Indexed: 01/12/2023]
Abstract
PURPOSE Nuclear respiratory factor 1 (NRF1) drives estrogen-dependent breast tumorigenesis. Herein we examined the impact of NRF1 activity on the aggressiveness and disparate molecular signature of breast cancer in Black, White, Asian, and Hispanic women. METHODS NRF1 activity by transcription factor target enrichment analysis and causal NRF1-target gene signatures by Bayesian Network Inference with Java Objects (BANJO) and Markov Chain Monte Carlo (MCMC)-based gene order were examined in The Cancer Genome Atlas (TCGA) breast cancer cohorts. RESULTS We are the first to report increased NRF1 activity based on its differential effects on genome-wide transcription associated with luminal A and B, HER2+ and triple-negative (TN) molecular subtypes of breast cancer in women of different race/ethnicity. We observed disparate NRF1 motif-containing causal gene signatures unique to Black, White, Asian, and Hispanic women for luminal A breast cancer. Further gene order searches showed molecular heterogeneity of each subtype of breast cancer. Six different gene order sequences involving CDK1, HMMR, CCNB2, CCNB1, E2F1, CREB3L4, GTSE1, and LMNB1 with almost equal weight predicted the probability of luminal A breast cancer in whites. Three different gene order sequences consisting of CCNB1 and GTSE1, and CCNB1, LMNB1, CDK1 or CASP3 predicted almost 100% probability of luminal B breast cancer in whites; CCNB1 and LMNB1 or GTSE predicted 100% HER2+ breast cancer in whites. GTSE1 and TUBA1C combined together predicted 100% probability of developing TNBC in whites; NRF1, TUBA1B and BAX with EFNA4, and NRF1 and BTRC predicated 100% TNBC in blacks. High expressor NRF1 TN breast tumors showed unfavorable prognosis with a high risk of breast cancer death in white women. CONCLUSION Our findings showed how sensitivity to high NRF1 transcriptional activity coupled with its target gene signatures contribute to racial differences in luminal A and TN breast cancer subtypes. This knowledge may be useful in personalized intervention to prevent and treat this clinically challenging problem.
Collapse
Affiliation(s)
- Jairo Ramos
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Changwon Yoo
- Department of Biostatistics, Florida International University, Miami, FL, 33199, USA
| | - Quentin Felty
- Department of Environmental Health Sciences, Florida International University, Miami, USA
| | - Zhenghua Gong
- Department of Biostatistics, Florida International University, Miami, FL, 33199, USA
| | - Juan P Liuzzi
- Department of Dietetics and Nutrition, Florida International University, Miami, FL, 33199, USA
| | - Robert Poppiti
- Department of Pathology, Florida International University, Miami, FL, USA
| | - Indu Shekhar Thakur
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ruchika Goel
- Medanta Cancer Institute, Medanta-The Medicity, Gurugram, Haryana, 122001, India
| | - Ashok Kumar Vaid
- Medanta Cancer Institute, Medanta-The Medicity, Gurugram, Haryana, 122001, India
| | - Ricardo Jorge Komotar
- Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, USA
| | - Nasreen Z Ehtesham
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Seyed E Hasnain
- JH Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Deodutta Roy
- Department of Environmental Health Sciences, Florida International University, Miami, USA.
| |
Collapse
|
19
|
Fei F, Liu K, Li C, Du J, Wei Z, Li B, Li Y, Zhang Y, Zhang S. Molecular Mechanisms by Which S100A4 Regulates the Migration and Invasion of PGCCs With Their Daughter Cells in Human Colorectal Cancer. Front Oncol 2020; 10:182. [PMID: 32154176 PMCID: PMC7047322 DOI: 10.3389/fonc.2020.00182] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/03/2020] [Indexed: 01/09/2023] Open
Abstract
Recently, an increasing number of evidences have shown that polyploid giant cancer cells (PGCCs) could generate daughter cells with a strong migration and invasion ability, which have been implicated in cancer recurrence and metastasis. However, the underlying molecular mechanisms of PGCCs with their daughter cells remain largely unclear. In vitro and in vivo experiments combined with 222 cases of human colorectal cancer (CRC) samples were used to identify the molecular mechanisms of S100A4-related proteins regulating the invasion and metastasis of PGCCs with their daughter cells. PGCCs with their daughter cells had high migration, invasion, and proliferation abilities compared to control cells; these were significantly inhibited after S100A4 knockdown. The high expression of cathepsin B, cyclin B1, TRIM21, and Annexin A2 were significantly downregulated after S100A4 knockdown, while the overexpression of S100A4, cathepsin B, cyclin B1, and S100A10 were significantly downregulated after TRIM21 knockdown in PGCCs with their daughter cells. The tumorigenic and metastatic ability of PGCCs with their daughter cells in vivo was significantly stronger compared to the untreated cells, which was significantly decreased after S100A4 knockdown. Moreover, the expression of S100A4-related proteins was positively correlated with the malignancy degree of human CRC, and maintained a high level in lymph node metastasis. S100A4 and TRIM21 may regulate each other to affect the expression and subcellular localization of cyclin B1, and participate in regulating the structure and function of Annexin A2/S100A10 complex, affecting downstream cathepsin B, resulting in the invasion and metastasis of PGCCs with their daughter cells. Besides, 14-3-3 ζ/δ and Ezrin may be involved in the motility and invasion of PGCCs with their daughter cells via cytoskeletal constructions with S100A4.
Collapse
Affiliation(s)
- Fei Fei
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China.,Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Kai Liu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Chunyuan Li
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Jiaxing Du
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhen Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bo Li
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuwei Li
- Departments of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Yi Zhang
- Departments of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
20
|
Han Y, Nakayama J, Hayashi Y, Jeong S, Futakuchi M, Ito E, Watanabe S, Semba K. Establishment and characterization of highly osteolytic luminal breast cancer cell lines by intracaudal arterial injection. Genes Cells 2020; 25:111-123. [PMID: 31849141 DOI: 10.1111/gtc.12743] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/18/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023]
Abstract
Bone is one of the most common metastatic sites of breast cancer, and bone metastasis profoundly affects the quality of life of breast cancer patients. Bone metastasis is commonly observed among all the subtypes of breast cancer; however, its molecular mechanism has been analyzed only in triple-negative subtype of breast cancer (TNBC). To characterize the molecular mechanisms of bone metastasis of luminal breast cancer, we established a bone-metastatic model of the MCF7, luminal breast cancer cell line, with enhanced osteolytic activity by intracaudal arterial injection (CAI). Pathological analysis of the established cell lines revealed that they exhibited fierce osteolytic ability by promoting osteoclast differentiation and activity. The signature genes extracted from highly osteolytic MCF7 cell lines were differed from those of bone-metastatic TNBC cell lines. Our results suggest that unique mechanisms of osteolysis in bone-metastatic lesions of luminal breast cancer. In addition, several up-regulated genes in MCF7-BM (Bone Metastasis) 02 cell lines correlated with poor prognosis with luminal breast cancer patients. Our findings support further study on the bone-metastatic mechanisms of luminal breast cancer.
Collapse
Affiliation(s)
- Yuxuan Han
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Jun Nakayama
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.,Computational Bio-Big Data Open Innovation Lab. (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Yusuke Hayashi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Seongmoon Jeong
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Mitsuru Futakuchi
- Department of Pathology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Emi Ito
- Department of Biomolecular Profiling, Translational Research Center, Fukushima Medical University, Fukushima, Japan
| | - Shinya Watanabe
- Department of Biomolecular Profiling, Translational Research Center, Fukushima Medical University, Fukushima, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.,Department of Cell Factory, Translational Research Center, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
21
|
Carmona S, Brunel JM, Bonier R, Sbarra V, Robert S, Borentain P, Lombardo D, Mas E, Gerolami R. A squalamine derivative, NV669, as a novel PTP1B inhibitor: in vitro and in vivo effects on pancreatic and hepatic tumor growth. Oncotarget 2019; 10:6651-6667. [PMID: 31803360 PMCID: PMC6877102 DOI: 10.18632/oncotarget.27286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 10/02/2019] [Indexed: 12/14/2022] Open
Abstract
NV669 is an aminosterol derived from squalamine found to possess strong anticancer effects. The aim of this study was to investigate NV669’s beneficial effects on human pancreatic and hepatic cancer models and to decipher the cellular and molecular mechanisms involved in tumor growth decrease upon treatment with NV669. Pancreatic (BxPC3, MiaPaCa-2) and hepatic (HepG2, Huh7) cancer cells were treated with NV669, and the effects recorded on proliferation, cell cycle and death. Results showed that NV669 inhibited the viability of cancer cells, induced cell cycle arrest and subsequently promoted apoptosis. This was accompanied by a decrease in the expression of cyclin B1 and phosphorylated Cdk1 and by a cleavage of pro-apoptotic caspase-8 and PARP-1. Taken together, our studies showed that NV669 inhibits the proliferation of pancreatic and hepatic cancer cells through the regulation of G2/M phase transition via the cyclin B1-Cdk1 complex. In vitro NV669 inhibits PTP1B activity and FAK expression. NV669 impacts on the expression of adhesion molecules CDH-1, -2 and -3 in BxPC3 and Huh7 lines that form cell monolayers. Consecutively NV669 induces cell detachment. This suggests that NV669 by inhibiting PTP1B induces cell detachment and apoptosis.
Subsequently, our in vivo results showed that NV669 inhibited the growth of pancreatic and hepatic tumor xenografts with a significant cell cycle arrest in pre-mitotic phase and an increase of tumor cell apoptosis. Therefore, NV669 may serve as an alternative anticancer agent, used alone or in association with other medications, for the treatment of pancreatic adenocarcinoma and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sylvie Carmona
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France.,Aix Marseille Univ, CNRS, INP, Institut de Neuro-Physiopathologie, Faculté de médecine, Marseille, France
| | | | - Rénaté Bonier
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France.,Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Véronique Sbarra
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Faculté de médecine, Marseille, France
| | - Stéphane Robert
- Aix Marseille Univ, INSERM, INRA, C2VN AMUTICYT Core facility, Faculté de pharmacie, Marseille, France
| | - Patrick Borentain
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France.,Aix Marseille Univ, AP-HM, Assistance Publique des Hôpitaux de Marseille, Centre Hospitalo-Universitaire Timone, Service d'Hépato-Gastro-Entérologie, Marseille, France
| | - Dominique Lombardo
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France
| | - Eric Mas
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France.,Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - René Gerolami
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de médecine, Marseille, France.,Aix Marseille Univ, AP-HM, Assistance Publique des Hôpitaux de Marseille, Centre Hospitalo-Universitaire Timone, Service d'Hépato-Gastro-Entérologie, Marseille, France
| |
Collapse
|
22
|
Shi Y, Li Y, Yan C, Su H, Ying K. Identification of key genes and evaluation of clinical outcomes in lung squamous cell carcinoma using integrated bioinformatics analysis. Oncol Lett 2019; 18:5859-5870. [PMID: 31788059 PMCID: PMC6865087 DOI: 10.3892/ol.2019.10933] [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: 05/12/2019] [Accepted: 09/02/2019] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Despite progress in the treatment of non-small-cell lung cancer, there are limited treatment options for lung squamous cell carcinoma (LUSC), compared with lung adenocarcinoma. The present study investigated the disease mechanism of LUSC in order to identify key candidate genes for diagnosis and therapy. A total of three gene expression profiles (GSE19188, GSE21933 and GSE74706) were analyzed using GEO2R to identify common differentially expressed genes (DEGs). The DEGs were then investigated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. A protein-protein interaction (PPI) network was constructed via the Search Tool for the Retrieval of Interacting Genes/Proteins, and visualized using Cytoscape software. The expression levels of the hub genes identified using CytoHubba were validated using the University of California, Santa Cruz (UCSC) database and the Human Protein Atlas. A Kaplan-Meier curve and Gene Expression Profiling Interactive Analysis were then employed to evaluate the associated prognosis and clinical pathological stage of the hub genes. Furthermore, non-coding RNA regulatory networks were constructed using the Gene-Cloud Biotechnology information website. A total of 359 common DEGs (155 upregulated and 204 downregulated) were identified, which were predominantly enriched in 'mitotic nuclear division', 'cell division', 'cell cycle' and 'p53 signaling pathway'. The PPI network consisted of 257 nodes and 2,772 edges, and the most significant module consisted of 66 upregulated genes. A total of 19 hub genes exhibited elevated RNA levels, and 10 hub genes had elevated protein levels compared with normal lung tissues. The upregulation of five hub genes (CCNB1, CEP55, FOXM1, MKI67 and TYMS; defined in Table I) were significantly associated with poor overall survival and unfavorable clinical pathological stages. Various ncRNAs, such as C1orf220, LINC01561 and MGC39584, may also play important roles in hub-gene regulation. In conclusion, the present study provides further understanding of the pathogenesis of LUSC, and reveals CCNB1, CEP55, FOXM1, MKI67 and TYMS as potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Yangfeng Shi
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Yeping Li
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Chao Yan
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Hua Su
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Kejing Ying
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
23
|
Li B, Zhu HB, Song GD, Cheng JH, Li CZ, Zhang YZ, Zhao P. Regulating the CCNB1 gene can affect cell proliferation and apoptosis in pituitary adenomas and activate epithelial-to-mesenchymal transition. Oncol Lett 2019; 18:4651-4658. [PMID: 31611974 PMCID: PMC6781518 DOI: 10.3892/ol.2019.10847] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the role and potential regulatory mechanisms of cyclin B1 (CCNB1) in the proliferation, apoptosis and epithelial-to-mesenchymal transition (EMT) in pituitary adenomas. A total of 24 specimens were included in the present study. The expression levels of CCNB1 protein in two normal pituitary and 22 pituitary adenoma tissues were determined by western blotting. CCNB1 was knocked-down by lentiviral-mediated infection of short hairpin RNA (shRNA) in GH3 and MMQ cell lines. The proliferation, cell cycle and apoptosis of GH3 and MMQ cell lines were detected using a Cell Counting Kit-8 and flow cytometer. Reverse transcription-quantitative PCR was utilized to detect the expression level of CCNB1 gene and EMT markers. In the present study, resveratrol (RES) was used as an inhibitor of CCNB1. The protein expression level of CCNB1 in pituitary adenomas was higher than that in normal pituitary tissue, as assessed by western blot analysis. In addition, the expression level of CCNB1 in invasive pituitary adenomas was higher when comparing invasive pituitary adenomas and non-invasive pituitary adenomas. Knockdown of CCNB1 resulted in significant decreases in cell viability and proliferation, arrested cell cycle at the G2/M phase and increased apoptosis. In addition, knockdown of CCNB1 significantly decreased the expression levels of the mesothelial cell marker N-cadherin (P<0.001), but significantly increased the expression levels of the epithelial cell markers E-cadherin (P<0.01) and p120-catenin (P<0.001). Further analyses identified that RES inhibited the expression level of CCNB1, and RES treatment exhibited a similar effect as CCNB1 shRNA infection. The present study suggested that suppressing the expression level of CCNB1 could regulate the proliferation and apoptosis of pituitary tumor cells and alter the expression level of various EMT markers. In addition, RES treatment could be used as an inhibitor of CCNB1. The present study also identified the molecular mechanisms underlying CCNB1 role in EMT.
Collapse
Affiliation(s)
- Bin Li
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Hai-Bo Zhu
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Gui-Dong Song
- Department of Cell and Biology, Beijing Neurosurgical Institute, Beijing 100070, P.R. China
| | - Jian-Hua Cheng
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Chu-Zhong Li
- Department of Cell and Biology, Beijing Neurosurgical Institute, Beijing 100070, P.R. China
| | - Ya-Zhuo Zhang
- Department of Cell and Biology, Beijing Neurosurgical Institute, Beijing 100070, P.R. China
| | - Peng Zhao
- Neurosurgical Department, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
24
|
Zhong G, Lou W, Yao M, Du C, Wei H, Fu P. Identification of novel mRNA-miRNA-lncRNA competing endogenous RNA network associated with prognosis of breast cancer. Epigenomics 2019; 11:1501-1518. [PMID: 31502865 DOI: 10.2217/epi-2019-0209] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To identify novel competing endogenous RNA (ceRNA) network related to patients prognosis in breast cancer. Materials & methods: Dysregulated mRNA based on intersection of three Gene Expression Omnibus and The Cancer Genome Atlas datasets were analyzed by bioinformatics. Results: In total 72 upregulated and 208 downregulated genes were identified. Functional analysis showed that some pathways related to cancer were significantly enriched. By means of stepwise reverse prediction and validation from mRNA to lncRNA, 19 hub genes, nine key miRNA and four key lncRNAs were identified by expression and survival analysis. Ultimately, the coexpression analysis identified RRM2-let-7a-5p-SNHG16/MAL2 as key ceRNA subnetwork associated with prognosis of breast cancer. Conclusion: We successfully constructed a novel ceRNA network, among which each component was significantly associated with breast cancer prognosis.
Collapse
Affiliation(s)
- Guansheng Zhong
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| | - Weiyang Lou
- Program of Innovative Cancer Therapeutics, Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, College of Medicine, Key Laboratory of Organ Transplantation, Zhejiang University, 79 Qingchun Road, Zhejiang Province, Hangzhou 310003, PR China
| | - Minya Yao
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| | - Chengyong Du
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| | - Haiyan Wei
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| |
Collapse
|
25
|
Liu J, Ma Z, Liu Y, Wu L, Hou Z, Li W. Screening of potential biomarkers in hepatitis C virus-induced hepatocellular carcinoma using bioinformatic analysis. Oncol Lett 2019; 18:2500-2508. [PMID: 31452738 PMCID: PMC6676667 DOI: 10.3892/ol.2019.10578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/06/2019] [Indexed: 01/10/2023] Open
Abstract
Evidence suggests that hepatitis C virus (HCV) infection is among the main causes of hepatocellular carcinoma (HCC). In addition, HCV-induced HCC (HCV-HCC) exhibits adverse clinical outcomes and limited therapeutic treatments are available for this condition. To investigate key biomarkers in the occurrence and development of HCV-HCC, microarray datasets GSE62232, GSE69715 and GSE107170 were downloaded from the Gene Expression Omnibus database for analysis. The differentially expressed genes between HCV-HCC and normal tissue were identified using the GEO2R online tool. The function enrichment analyses including Gene Ontology and Kyoto Encyclopedia of Genes and Genomes were performed using the Database for Annotation, Visualization and Integrated Discovery online tool. A protein-protein interaction network was constructed using the Search Tool for the Retrieval of Interacting Genes database and visualized using Cytoscape. A total of 368 DEGs were identified, and the top 10 hub genes with a high degree of connectivity were selected for further analysis. Subsequently, overall survival and disease-free survival analysis revealed that there was a significant association between altered expression of HMMR, CCNB1 and KIF20A, and poor clinical outcome. In summary, these results indicate that HMMR, CCNB1 and KIF20A are potential targets for diagnosis and therapy of HCV-HCC.
Collapse
Affiliation(s)
- Jun Liu
- Department of Laboratory Medicine, Yue Bei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Zhanzhong Ma
- Department of Laboratory Medicine, Yue Bei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Yanming Liu
- Department of Laboratory Medicine, Yue Bei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Liangyin Wu
- Department of Laboratory Medicine, Yue Bei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Zhiwei Hou
- Reproductive Medicine Center, Yue Bei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| | - Wenli Li
- Reproductive Medicine Center, Yue Bei People's Hospital, Shaoguan, Guangdong 512026, P.R. China
| |
Collapse
|
26
|
Li X, Nie C, Tian B, Tan X, Han W, Wang J, Jin Y, Li Y, Guan X, Hong A, Chen X. miR-671-5p Blocks The Progression Of Human Esophageal Squamous Cell Carcinoma By Suppressing FGFR2. Int J Biol Sci 2019; 15:1892-1904. [PMID: 31523191 PMCID: PMC6743296 DOI: 10.7150/ijbs.32429] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/02/2019] [Indexed: 12/31/2022] Open
Abstract
Esophageal cancer is the eighth most common malignant tumor worldwide, of which esophageal squamous cell carcinoma (ESCC) is the dominant histological subtype. A drug shortage for ESCC therapy triggered us to explore the roles of fibroblast growth factor receptor 2 (FGFR2) and its upstream regulator miR-671-5p in ESCC progression. We compared the levels of FGFR2 and miR-671-5p between human ESCC tissues and their matched normal esophageal tissues and found an association between higher levels of FGFR2 and lower levels of miR-671-5p in ESCC tissues. High levels of FGFR2 resulted in the activation of the ERK and AKT pathways and a promotion of ESCC progression. High levels of miR-671-5p specifically reduced the expression of FGFR2 and suppressed ESCC progression in both in vitro and in vivo models. Therefore, suppressing FGFR2 and enhancing miR-671-5p expression may be the right approaches for ESCC therapy.
Collapse
Affiliation(s)
- Xiaoyan Li
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Changjun Nie
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Baoqing Tian
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xuan Tan
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Wei Han
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Jiakang Wang
- Cancer Center of Guangzhou Medical University, Guangzhou, Guangdong, 510090, P. R. China
| | - Yuan Jin
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Yadan Li
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong, China
| | - An Hong
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| | - Xiaojia Chen
- Institute of Biomedicine & Department of cell Biology, Jinan University, Guangzhou, Guangdong, 510632, P. R. China.,National Engineering Research Center of Genetic Medicine, Guangzhou, Guangdong, 510632, P. R. China.,Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou, Guangdong, 510632, P. R. China
| |
Collapse
|
27
|
Khumukcham SS, Samanthapudi VSK, Penugurti V, Kumari A, Kesavan PS, Velatooru LR, Kotla SR, Mazumder A, Manavathi B. Hematopoietic PBX-interacting protein is a substrate and an inhibitor of the APC/C-Cdc20 complex and regulates mitosis by stabilizing cyclin B1. J Biol Chem 2019; 294:10236-10252. [PMID: 31101654 DOI: 10.1074/jbc.ra118.006733] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/27/2019] [Indexed: 02/04/2023] Open
Abstract
Proper cell division relies on the coordinated regulation between a structural component, the mitotic spindle, and a regulatory component, anaphase-promoting complex/cyclosome (APC/C). Hematopoietic PBX-interacting protein (HPIP) is a microtubule-associated protein that plays a pivotal role in cell proliferation, cell migration, and tumor metastasis. Here, using HEK293T and HeLa cells, along with immunoprecipitation and immunoblotting, live-cell imaging, and protein-stability assays, we report that HPIP expression oscillates throughout the cell cycle and that its depletion delays cell division. We noted that by utilizing its D box and IR domain, HPIP plays a dual role both as a substrate and inhibitor, respectively, of the APC/C complex. We observed that HPIP enhances the G2/M transition of the cell cycle by transiently stabilizing cyclin B1 by preventing APC/C-Cdc20-mediated degradation, thereby ensuring timely mitotic entry. We also uncovered that HPIP associates with the mitotic spindle and that its depletion leads to the formation of multiple mitotic spindles and chromosomal abnormalities, results in defects in cytokinesis, and delays mitotic exit. Our findings uncover HPIP as both a substrate and an inhibitor of APC/C-Cdc20 that maintains the temporal stability of cyclin B1 during the G2/M transition and thereby controls mitosis and cell division.
Collapse
Affiliation(s)
| | | | - Vasudevarao Penugurti
- From the Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India and
| | - Anita Kumari
- From the Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India and
| | - P S Kesavan
- the Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, Hyderabad 500107, Telangana, India
| | - Loka Reddy Velatooru
- From the Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India and
| | - Siva Reddy Kotla
- From the Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India and
| | - Aprotim Mazumder
- the Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, Hyderabad 500107, Telangana, India
| | - Bramanandam Manavathi
- From the Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India and
| |
Collapse
|
28
|
Prediction of onset of remnant gastric cancer by promoter DNA methylation of CDO1/ HOPX/ Reprimo/ E-cadherin. Oncotarget 2019; 10:2423-2434. [PMID: 31069006 PMCID: PMC6497431 DOI: 10.18632/oncotarget.26814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background Early detection of remnant gastric cancer (RGC) is required to reduce the risk of death, but long-term endoscopic surveillance is difficult after gastrectomy. In this study, data for the methylation status of 4 methylation genes (CDO1, HOPX, Reprimo, and E-cadherin) to predict the onset of RGC are presented. Results The 4 genes showed hypermethylation in RGC tumors in contrast to the corresponding non-cancerous mucosa tissues. The methylation level in the non-cancerous mucosa tissues of the initial surgery was obviously high in initial malignant disease for CDO1 (P = 0.0001), while in initial benign one for E-cadherin (P = 0.003). Promoter DNA methylation status in the remnant non-cancerous mucosa tissues together with the basic clinical data in turn predicted either initial malignant disease or initial benign disease with a high AUC score of 0.94, suggesting that methylation events are differentially recognized between the initial malignant and benign disease. We then finally confirmed that 4 genes hypermethylation of the non-cancerous tissues by biopsy prior to onset of RGC could predict terms until RGC occurred (P < 0.0001). Methods A total of 58 RGC patients were used to establish the model. The 4 genes promoter methylation were analyzed for DNA obtained from the patient's specimens using quantitative methylation specific polymerase chain reaction. Conclusions This risk model would help provide guidance for endoscopic surveillance plan of RGC after gastrectomy.
Collapse
|
29
|
Sun Z, Li Y, Wang H, Cai M, Gao S, Liu J, Tong L, Hu Z, Wang Y, Wang K, Zhang L, Cao X, Zhang S, Shi F, Zhao J. miR-181c-5p mediates simulated microgravity-induced impaired osteoblast proliferation by promoting cell cycle arrested in the G 2 phase. J Cell Mol Med 2019; 23:3302-3316. [PMID: 30761733 PMCID: PMC6484313 DOI: 10.1111/jcmm.14220] [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: 10/21/2018] [Revised: 01/06/2019] [Accepted: 01/23/2019] [Indexed: 12/15/2022] Open
Abstract
Impaired osteoblast proliferation plays fundamental roles in microgravity‐induced bone loss, and cell cycle imbalance may result in abnormal osteoblast proliferation. However, whether microgravity exerts an influence on the cell cycle in osteoblasts or what mechanisms may underlie such an effect remains to be fully elucidated. Herein, we confirmed that simulated microgravity inhibits osteoblast proliferation. Then, we investigated the effect of mechanical unloading on the osteoblast cell cycle and found that simulated microgravity arrested the osteoblast cell cycle in the G2 phase. In addition, our data showed that cell cycle arrest in osteoblasts from simulated microgravity was mainly because of decreased cyclin B1 expression. Furthermore, miR‐181c‐5p directly inhibited cyclin B1 protein translation by binding to a target site in the 3′UTR. Lastly, we demonstrated that inhibition of miR‐181c‐5p partially counteracted cell cycle arrest and decreased the osteoblast proliferation induced by simulated microgravity. In conclusion, our study demonstrates that simulated microgravity inhibits cell proliferation and induces cell cycle arrest in the G2 phase in primary mouse osteoblasts partially through the miR‐181c‐5p/cyclin B1 pathway. This work may provide a novel mechanism of microgravity‐induced detrimental effects on osteoblasts and offer a new avenue to further investigate bone loss induced by mechanical unloading.
Collapse
Affiliation(s)
- Zhongyang Sun
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China.,The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Ying Li
- Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Han Wang
- Department of Orthopedics, Affiliated Hospital of Air Force Aviation Medicine Research Institute, Fourth Military Medical University, Beijing, China
| | - Min Cai
- Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China.,Medical Services Section, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Shanshan Gao
- Medical Services Section, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Jing Liu
- Department of Pharmacy, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Liangcheng Tong
- Department of Orthopedics, Junxie Hospital, Anhui Medical University, Nanjing, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Yixuan Wang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Ke Wang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Lijun Zhang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Xinsheng Cao
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Chinese Ministry of Education, Fourth Military Medical University, Xi'an, China
| | - Jianning Zhao
- Department of Orthopedics, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
30
|
Yan Z, Liu G, Liang M, Xu Y. Ophiopogonin D inhibits cell proliferation and induces apoptosis of human laryngocarcinoma through downregulation of cyclin B1 and MMP-9 and upregulation of p38-MAPK signaling. Oncol Lett 2019; 17:1877-1882. [PMID: 30675250 DOI: 10.3892/ol.2018.9788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 11/06/2017] [Indexed: 11/06/2022] Open
Abstract
The pharmacological actions of Ophiopogonin D include resistance to cardiovascular and cerebrovascular diseases, anti-aging effects, improvement in learning deficit and dysmnesia, anti-tumor, anti-radiation and anti-inflammatory effects, immunoregulation, and the relief of cough and hepatopulmonary pathological lesions. However, the efficacy of Ophiopogonin D on human laryngocarcinoma remains to be elucidated. The present study therefore investigated whether the anti-cancer effect of Ophiopogonin D inhibits cell proliferation and induces apoptosis of human laryngocarcinoma. In the present study, it was found that Ophiopogonin D inhibited cell proliferation, promoted cytotoxicity, induced apoptosis and increased caspase-3/9 activity in human laryngocarcinoma cells. Ophiopogonin D significantly suppressed cyclin B1 and matrix metalloproteinase-9 (MMP-9) protein expression, and upregulated p-p38 MAPK protein expression in human laryngocarcinoma cells. Together, these results suggest Ophiopogonin D inhibits cell proliferation and induced apoptosis in human laryngocarcinoma cells through downregulation of cyclin B1 and MMP-9 and upregulation of the p38 MAPK signaling pathway. Therefore, Ophiopogonin D may be a potential therapy for the treatment of human laryngocarcinoma.
Collapse
Affiliation(s)
- Zhiyu Yan
- Department of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| | - Guang Liu
- Department of Pathology, Beijing Military General Hospital, Beijing 100700, P.R. China
| | - Min Liang
- Department of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| | - Yanjun Xu
- Department of Otolaryngology, Beijing Military General Hospital, Beijing 100700, P.R. China
| |
Collapse
|
31
|
Upregulated expression of HOXB7 in intrahepatic cholangiocarcinoma is associated with tumor cell metastasis and poor prognosis. J Transl Med 2019; 99:736-748. [PMID: 30664713 PMCID: PMC6760572 DOI: 10.1038/s41374-018-0150-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023] Open
Abstract
Homeobox B7 (HOXB7) protein is reported to be aberrantly expressed in a variety of cancers and to play an important role in multiple cellular processes. However, the specific mechanism by which HOXB7 promotes the malignant progression of intrahepatic cholangiocarcinoma (ICC) remains unclear. Therefore, we used quantitative real-time polymerase chain reaction (PCR) to detect the expression level of HOXB7 in 38 paired ICC tissue samples. Additionally, to assess correlation between HOXB7 and ICC prognosis, we performed immunohistochemistry (IHC) using 122 ICC tissues to detect HOXB7 expression. Cell Counting Kit-8 (CCK-8) and colony formation assays were employed to assess ICC cell proliferation, and Transwell assays were performed to estimate the invasion and migration abilities of ICC cells. The capillary tube formation assay was applied to explore the angiogenic effects of HOXB7. A xenograft tumor model was established in nude mice to assess the role of HOXB7 in tumor growth and lung metastasis. The results showed higher expression of HOXB7 in ICC tissues than in noncancerous tissues, and this increased expression was significantly associated with a poor prognosis. In addition, HOXB7 overexpression enhanced capillary tube formation, invasion and migration of ICC cells in vitro, whereas HOXB7 knockdown produced the opposite results in vitro. Moreover, the role of HOXB7 in promoting tumor growth and metastasis was verified in vivo. Further investigation revealed that the expression levels of MMP2, MMP9, VEGFa, and IL8 were elevated by HOXB7 and that the ERK pathway was activated. Our results demonstrate the prognostic value of HOXB7 and its role in metastasis and angiogenesis in ICC. HOXB7 upregulated MMP2, MMP9, VEGFa, and IL8 expression via the ERK pathway to accelerate the malignant progression of ICC.
Collapse
|
32
|
Gong L, Zhang D, Dong Y, Lei Y, Qian Y, Tan X, Han S, Wang J. Integrated Bioinformatics Analysis for Identificating the Therapeutic Targets of Aspirin in Small Cell Lung Cancer. J Biomed Inform 2018; 88:20-28. [PMID: 30414472 DOI: 10.1016/j.jbi.2018.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/25/2018] [Accepted: 11/05/2018] [Indexed: 01/21/2023]
Abstract
PURPOSE We explored the mechanism of aspirin in SCLC by dissecting many publicly available databases. METHODS AND RESULTS Firstly, 11 direct protein targets (DPTs) of aspirin were identified by DrugBank 5.0. Then protein-protein interaction (PPI) network and signaling pathways of aspirin DPTs were analyzed. We found that aspirin was linked with many kinds of cancer, and the most significant one is SCLC. Next, we classified the mutation of 4 aspirin DPTs in SCLC (IKBKB, NFKBIA, PTGS2 and TP53) using cBio Portal. Further, we identified top 50 overexpressed genes of SCLC by Oncomine, and the interconnected genes with the 4 aspirin DPTs in SCLC (IKBKB, NFKBIA, PTGS2 and TP53) by STRING. Lastly, we figured out 5 consistently genes as potential therapeutic targets of aspirin in SCLC. CONCLUSION The integrated bioinformatical analysis could improve our understanding of the underlying molecular mechanism about how aspirin working in SCLC. Integrated bioinformatical analysis may be considered as a new paradigm for guiding future studies about interaction in drugs and diseases.
Collapse
Affiliation(s)
- Liuyun Gong
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Dan Zhang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, PR China
| | - Yiping Dong
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Yutiantian Lei
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Yuanjie Qian
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Xinyue Tan
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Suxia Han
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Jiquan Wang
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
33
|
Vicente C, Stirparo R, Demeyer S, de Bock CE, Gielen O, Atkins M, Yan J, Halder G, Hassan BA, Cools J. The CCR4-NOT complex is a tumor suppressor in Drosophila melanogaster eye cancer models. J Hematol Oncol 2018; 11:108. [PMID: 30144809 PMCID: PMC6109294 DOI: 10.1186/s13045-018-0650-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/13/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The CNOT3 protein is a subunit of the CCR4-NOT complex, which is involved in mRNA degradation. We recently identified CNOT3 loss-of-function mutations in patients with T-cell acute lymphoblastic leukemia (T-ALL). METHODS Here, we use different Drosophila melanogaster eye cancer models to study the potential tumor suppressor function of Not3, the CNOT3 orthologue, and other members of the CCR4-NOT complex. RESULTS Our data show that knockdown of Not3, the structural components Not1/Not2, and the deadenylases twin/Pop2 all result in increased tumor formation. In addition, overexpression of Not3 could reduce tumor formation. Not3 downregulation has a mild but broad effect on gene expression and leads to increased levels of genes involved in DNA replication and ribosome biogenesis. CycB upregulation also contributes to the Not3 tumor phenotype. Similar findings were obtained in human T-ALL cell lines, pointing out the conserved function of Not3. CONCLUSIONS Together, our data establish a critical role for Not3 and the entire CCR4-NOT complex as tumor suppressor.
Collapse
Affiliation(s)
- Carmen Vicente
- Center for Cancer Biology, VIB, Leuven, Belgium. .,Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium. .,Centro de Investigación Médica Aplicada, Av. de Pío XII, 55, 31008, Pamplona, Spain.
| | - Rocco Stirparo
- Center for Cancer Biology, VIB, Leuven, Belgium.,Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium
| | - Sofie Demeyer
- Center for Cancer Biology, VIB, Leuven, Belgium.,Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium
| | - Charles E de Bock
- Center for Cancer Biology, VIB, Leuven, Belgium.,Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium
| | - Olga Gielen
- Center for Cancer Biology, VIB, Leuven, Belgium.,Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium
| | - Mardelle Atkins
- Center for Cancer Biology, VIB, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jiekun Yan
- Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium.,Center for Brain & Disease Research, VIB, Leuven, Belgium
| | - Georg Halder
- Center for Cancer Biology, VIB, Leuven, Belgium.,Department of Oncology, KU Leuven, Leuven, Belgium
| | - Bassem A Hassan
- Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium.,Center for Brain & Disease Research, VIB, Leuven, Belgium.,Institut du Cerveau et de la Moelle Epinière (ICM) - Hôpital Pitié-Salpêtrière, UPMC, Sorbonne Universités, Inserm, CNRS, Paris, France
| | - Jan Cools
- Center for Cancer Biology, VIB, Leuven, Belgium. .,Center for Human Genetics, KU Leuven, Herestraat 49, box 912, B-3000, Leuven, Belgium.
| |
Collapse
|
34
|
Li MX, Jin LT, Wang TJ, Feng YJ, Pan CP, Zhao DM, Shao J. Identification of potential core genes in triple negative breast cancer using bioinformatics analysis. Onco Targets Ther 2018; 11:4105-4112. [PMID: 30140156 PMCID: PMC6054764 DOI: 10.2147/ott.s166567] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor clinical outcome and limited treatment options. Lacking molecular targets, chemotherapy is the main adjuvant treatment for TNBC patients. Materials and methods To explore potential therapeutic targets for TNBC, we analyzed three microarray datasets (GSE38959, GSE45827, and GSE65194) derived from the Gene Expression Omnibus (GEO) database. The GEO2R tool was used to screen out differentially expressed genes (DEGs) between TNBC and normal tissue. Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were performed using the Database for Annotation, Visualization and Integrated Discovery to identify the pathways and functional annotation of DEGs. Protein–protein interaction of these DEGs was analyzed based on the Search Tool for the Retrieval of Interacting Genes database and visualized by Cytoscape software. In addition, we used the online Kaplan–Meier plotter survival analysis tool to evaluate the prognostic value of hub genes expression in breast cancer patients. Results A total of 278 upregulated DEGs and 173 downregulated DEGs were identified. Among them, ten hub genes with a high degree of connectivity were picked out. Overexpression of these hub genes was associated with unfavorable prognosis of breast cancer, especially, CCNB1 overexpression was observed and indicated poor outcome of TNBC. Conclusion Our study suggests that CCNB1 was overexpressed in TNBC compared with normal breast tissue, and overexpression of CCNB1 was an unfavorable prognostic factor of TNBC patients. Further study is needed to explore the value of CCNB1 in the treatment of TNBC.
Collapse
Affiliation(s)
- Man-Xiu Li
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| | - Li-Ting Jin
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| | - Tie-Jun Wang
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| | - Yao-Jun Feng
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| | - Cui-Ping Pan
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| | - Dei-Mian Zhao
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| | - Jun Shao
- Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, People's Republic of China,
| |
Collapse
|
35
|
Ye C, Wang J, Wu P, Li X, Chai Y. Prognostic role of cyclin B1 in solid tumors: a meta-analysis. Oncotarget 2018; 8:2224-2232. [PMID: 27903976 PMCID: PMC5356794 DOI: 10.18632/oncotarget.13653] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/16/2016] [Indexed: 01/11/2023] Open
Abstract
Cyclin B1 is a key mitotic cyclin in the G2-M phase transition of the cell cycle and is overexpressed in various malignant tumors. Numerous studies have reported contradictory evidences of the correlation between cyclin B1 expression and prognosis in human solid tumors. To address this discrepancy, we conducted a meta-analysis with 17 published studies searched from PubMed and Medline. Cyclin B1 overexpression was significantly associated with poor 3-year overall survival (OS) (OR = 2.05, 95% CI = 1.20 to 3.50, P = 0.009) and 5-year OS (OR = 2.11, 95% CI = 1.33 to 3.36, P = 0.002) of solid tumors. Subgroup analysis revealed that elevated cyclin B1 expression was associated with worse prognosis of lung cancer and esophageal cancer but better prognosis of colorectal cancer. In summary, overexpression of cyclin B1 is correlated with poor survival in most solid tumors, which suggests that the expression status of cyclin B1 is a significant prognostic parameter in solid tumors.
Collapse
Affiliation(s)
- Chenyang Ye
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Ji Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Pin Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiaofen Li
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences, College of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Ying Chai
- Department of Thoracic Surgery, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
36
|
Xu W, Xia H, Liu W, Zheng W, Hua L. Exploration of genetics commonness between bladder cancer and breast cancer based on a silcio analysis on disease subtypes. Technol Health Care 2018; 26:361-377. [PMID: 29758961 PMCID: PMC6027900 DOI: 10.3233/thc-174699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Muscle-invasive bladder cancers (MIBCs) are heterogeneous cancers and can be grouped into basal-like and luminal subtypes that are highly reminiscent of those found in breast cancer. Like basal-like breast cancers, basal-like MIBCs are associated with advanced stage and metastatic disease. However, the biological and clinical significance of molecular subtypes of MIBCs remain unclear. Therefore, we implemented a serious of bioinformatics methods to explore genetic similarities between bladder and breast cancers. METHODS AND RESULTS In the current study, by the application of multiple levels data analysis including random forest analysis, PPI and transcription factor regulation network construction, Gene Ontology (GO) and KEGG pathway enrichment analysis, we explored the genetics commonness between MIBC and breast cancers from the molecular heterogeneity based on the disease subtypes. CONCLUSIONS Our study identified some basal-related and luminal-related genes shared by two cancers. These studies can help shed light on the potential relationships between MIBC and breast cancer as a whole.
Collapse
Affiliation(s)
- Wenbin Xu
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing 100069, China
| | - Hong Xia
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing 100069, China
| | - Wei Liu
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Weiying Zheng
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing 100069, China
| | - Lin Hua
- School of Biomedical Engineering, Capital Medical University, Beijing 100069, China
- Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, Beijing 100069, China
| |
Collapse
|
37
|
Shi Q, Wang W, Jia Z, Chen P, Ma K, Zhou C. ISL1, a novel regulator of CCNB1, CCNB2 and c-MYC genes, promotes gastric cancer cell proliferation and tumor growth. Oncotarget 2017; 7:36489-36500. [PMID: 27183908 PMCID: PMC5095015 DOI: 10.18632/oncotarget.9269] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 04/22/2016] [Indexed: 12/22/2022] Open
Abstract
Islet-1 (ISL1) belongs to the LIM homeodomain transcription factor family, which is specifically expressed in certain tissue types only. Previously, we reported that ISL1 is aberrantly overexpressed in gastric cancer (GC). However, its role in GC is not clear. Here, we report that ISL1 is aberrantly upregulated not only in human gastric carcinoma tissues but also in some GC cell lines. Upregulated ISL1 expression enhanced xenografted gastric carcinoma development, while ISL1 knockdown inhibited GC growth in nude mice. ISL1 overexpression promoted GC cell proliferation, colony formation, and cell growth in soft agar, and facilitated cell cycle transition in GC cells, demonstrated an increase in the proportion of cells in the G2/M and S phases and a decrease in the proportion of cells in the G1 phase. Furthermore, we provide evidence that ISL1 is a novel regulator of the cyclin B1 (CCNB1), cyclin B2 (CCNB2) and c-myc (c-MYC) genes. ISL1 activated the expression of these genes in GC cells by binding to the conserved binding sites on their promoters or enhancers. The expression levels of the genes were decreased in response to ISL1 knockdown. Therefore, ISL1 may serve as a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Qiong Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, Beijing, P.R. China
| | - Weiping Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, Beijing, P.R. China
| | - Zhuqing Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, Beijing, P.R. China
| | - Ping Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, Beijing, P.R. China
| | - Kangtao Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, Beijing, P.R. China
| | - Chunyan Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education of China, Peking University, Beijing, P.R. China
| |
Collapse
|
38
|
Wen Y, Cao L, Lian WP, Li GX. The prognostic significance of high/positive expression of cyclin B1 in patients with three common digestive cancers: a systematic review and meta-analysis. Oncotarget 2017; 8:96373-96383. [PMID: 29221213 PMCID: PMC5707107 DOI: 10.18632/oncotarget.21273] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 09/08/2017] [Indexed: 01/19/2023] Open
Abstract
Recently, several studies have reported that the expression of cyclin B1 may be associated with the prognosis of cancer. Nevertheless, their conclusions were still controversial. The present was designed to analyze and evaluate the prognostic role of cyclin B1 expression in patients with digestive cancer. PubMed, Embase, Cochrane Library and Web of Science were searched to January, 2017. Pooled odds ratio (OR) with 95% confidence intervals (CIs) were estimated. For the pooled OR estimates of OS, we performed subgroup analysis. Besides, sensitivity analysis was performed to examine the stability of the combined results. All statistical analyses were performed using standard statistical procedures provided in RevMan 5.2. A total of 12 studies (N = 2080 participants) were included for this meta-analysis. The positive/high expression of cyclin B1 had an obvious association with both 3-year overall survival (OR 0.21, 95% CI 0.12-0.37; P < 0.00001) and 5-year overall survival (OR 0.20, 95% CI 0.12-0.34; P < 0.00001) in esophageal cancer, and 5-year overall survival of colorectal cancer (OR 2.01, 95% CI 1.32-3.08; P = 0.001). This meta-analysis indicated that positive/high expression of cyclin B1 may have a close association with worse survival in patients with esophageal cancer, but better prognosis in patients with colorectal cancer.
Collapse
Affiliation(s)
- Yong Wen
- Chinese Medicine Department, Southwest Medical University Affiliated Hospital, Luzhou 646000, China
| | - Lei Cao
- Department of Pediatrics, Gansu Provincial Maternity and Child Care Hospital, Lanzhou 730000, China
| | - Wen-Ping Lian
- Department of Clinical Laboratory, The Third People's Hospital of He'nan Province, Zhengzhou 450000, China
| | - Guo-Xia Li
- Department of Pathology, Shanghai Minhang District Central Hospital, Shanghai 201199, China
| |
Collapse
|
39
|
Shao D, Ma J, Zhou C, Zhao JN, Li LL, Zhao TJ, Ai XL, Jiao P. STAT3 down-regulation induces mitochondria-dependent G2/M cell cycle arrest and apoptosis in oesophageal carcinoma cells. Clin Exp Pharmacol Physiol 2017; 44:413-420. [PMID: 27896845 DOI: 10.1111/1440-1681.12708] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 11/25/2016] [Accepted: 11/25/2016] [Indexed: 12/21/2022]
Abstract
STAT3 is persistently activated in a wide variety of human tumours, and aberrant STAT3 activity promotes tumour growth, invasion and metastasis. To explore STAT3 down-regulation in human oesophageal cancer cells, cell proliferation, apoptosis and mitochondrial mechanisms were explored in oesophageal carcinoma TE1 cell cultures. We demonstrate for the first time that STAT3 down-regulation by RNAi is sufficient to inhibit oesophageal cancer cell proliferation inducing cell apoptosis. Further, we demonstrate that mitochondrial transmembrane potential is impaired thereby leading to collapsed mitochondrial membrane potential, abnormal mitochondrial membrane depolarization, nuclear DNA fragmentation and cell cycle G2/M arrest under the conditions of STAT3 down-regulation. Thus, our results suggest that STAT3 inhibition is a valid approach to induce oesophageal carcinoma cell mitochondrial-dependent apoptosis in therapeutic strategies against oesophageal cancers.
Collapse
Affiliation(s)
- Dan Shao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China.,The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jie Ma
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Chao Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jia-Nan Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Lu-Lu Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Tong-Jian Zhao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xi-Lei Ai
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Ping Jiao
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
40
|
Boess F, Lenz B, Funk J, Niederhauser U, Bassett S, Zhang JD, Singer T, Roth AB. Use of early phenotypic in vivo markers to assess human relevance of an unusual rodent non-genotoxic carcinogen in vitro. Toxicology 2017; 379:48-61. [PMID: 28174063 DOI: 10.1016/j.tox.2017.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 10/20/2022]
Abstract
Foci of altered hepatocytes (FAH) are considered putative, pre-neoplastic lesions that can occur spontaneously in aging rodents, but can also be induced by chemicals or drugs. Progression of FAH to hepatocellular neoplasms has been reported repeatedly but increases in foci in rodents do not necessarily lead to tumors in carcinogenicity studies and the relevance for humans often remains unclear. Here we present the case of RG3487, a molecule which induced FAH and, later on, tumors in rats. Because the molecule was negative in genotoxicity assays it was classified as a non-genotoxic carcinogen. In order to assess the potential for liver tumor formation in humans, we analyzed treatment-induced changes in vivo to establish a possible mode of action (MoA). In vivo and in vitro gene expression analysis revealed that nuclear receptor signaling was unlikely to be the relevant MoA and no other known mechanism could be established. We therefore took an approach comparing phenotypic markers, including mRNA changes, proliferation and glycogen accumulation, in vitro using cells of different species to assess the human relevance of this finding. Since the alterations observed in rats were not seen in the liver of mice or dogs in vivo, we could validate the relevance of the cell models chosen by use of hepatocytes from these species in vitro. This ultimately allowed for a cross-species comparison, which suggested that the formation of FAH and liver tumors was rat specific and unlikely to translate to human. Our work showed that phenotypic species comparison in vitro is a useful approach for assessment of the human relevance of pre-clinical findings where no known mechanism can be established.
Collapse
Affiliation(s)
- Franziska Boess
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Barbara Lenz
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Juergen Funk
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Urs Niederhauser
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Simon Bassett
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Jitao David Zhang
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Thomas Singer
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | - Adrian B Roth
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| |
Collapse
|
41
|
Yao M, Shang YY, Zhou ZW, Yang YX, Wu YS, Guan LF, Wang XY, Zhou SF, Wei X. The research on lapatinib in autophagy, cell cycle arrest and epithelial to mesenchymal transition via Wnt/ErK/PI3K-AKT signaling pathway in human cutaneous squamous cell carcinoma. J Cancer 2017; 8:220-226. [PMID: 28243326 PMCID: PMC5327371 DOI: 10.7150/jca.16850] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/10/2016] [Indexed: 01/09/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) contributes to one of most common types of skin cancer. Epidermal growth factor receptor (EGFR) activation has been investigated to be associated with the development of cSCC. Lapatinib is an inhibitor targeting HER2/neu and EGFR pathway. We found that lapatinib can inhibit proliferation by enhancing apoptosis of human cSCC cell lines. The cSCC cell cycle distribution could be arrested in G2/M phase after lapatinib treatment. In the in vitro experiment, we found that lapatinib interrupted PI3K/AKT/mTOR signaling pathway in human cSCC cells. Furthermore, lapatinib could suppress epithelial to mesenchymal transition (EMT) via Wnt/ErK/PI3K-AKT signaling pathway to represent a promising anticancer drug for cSCC treatment.
Collapse
Affiliation(s)
- Ming Yao
- Department of Burns and Plastic Surgery, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China.; Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yuan-Yuan Shang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA.; Department of Dermatology, General Hospital, Ningxia Medical University
| | - Zhi-Wei Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yin-Xue Yang
- Department of Colorectal Surgery, General Hospital, Ningxia Medical University
| | - Yin-Sheng Wu
- Department of Burns and Plastic Surgery, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Li-Feng Guan
- Department of Burns and Plastic Surgery, General Hospital, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Xin-Yu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA.; Department of Pharmacy, General Hospital, Ningxia Medical University, Yinchuan
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
42
|
Bozok Cetintas V, Acikgoz E, Yigitturk G, Demir K, Oktem G, Tezcanli Kaymaz B, Oltulu F, Aktug H. Effects of flavopiridol on critical regulation pathways of CD133high/CD44high lung cancer stem cells. Medicine (Baltimore) 2016; 95:e5150. [PMID: 27787370 PMCID: PMC5089099 DOI: 10.1097/md.0000000000005150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Flavopiridol a semisynthetic flavone that inhibits cyclin-dependent kinases (CDKs) and has growth-inhibitory activity and induces a blockade of cell-cycle progression at G1-phase and apoptosis in numerous human tumor cell lines and is currently under investigation in phase II clinical trials. Cancer stem cells (CSCs) are comprised of subpopulation of cells in tumors that have been proposed to be responsible for recurrence and resistance to chemotherapy. The aim of the present study was to investigate the effects of flavopiridol in cancer stem cell cytoskeleton, cell adhesion, and epithelial to mesenchymal transition in CSCs. METHODS The cells were treated with flavopiridol to determine the inhibitory effect. Cell viability and proliferation were determined by using the WST-1 assay. Caspase activity and immunofluorescence analyses were performed for the evaluation of apoptosis, cell cytoskeleton, and epithelial-mesenchymal transition (EMT) markers. The effects of flavopiridol on the cell cycle were also evaluated. Flow cytometric analysis was used to detect the percentages of CSCs subpopulation. We analyzed the gene expression patterns to predict cell cycle and cell cytoskeleton in CSCs by RT-PCR. RESULTS Flavopiridol-induced cytotoxicity and apoptosis at the IC50 dose, resulting in a significant increase expression of caspases activity. Cell cycle analyses revealed that flavopiridol induces G1 phase cell cycle arrest. Flavopiridol significantly decreased the mRNA expressions of the genes that regulate the cell cytoskeleton and cell cycle components and cell motility in CSCs. CONCLUSION Our results suggest that Flavopiridol has activity against lung CSCs and may be effective chemotherapeutic molecule for lung cancer treatment.
Collapse
Affiliation(s)
| | - Eda Acikgoz
- Department of Histology and Embryology, Yuzuncu Yil University Faculty of Medicine, Van
- Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey
- Correspondence: Eda Acikgoz, Yuzuncu Yil University Faculty of Medicine, Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey (e-mail: )
| | - Gurkan Yigitturk
- Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Kenan Demir
- Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Gulperi Oktem
- Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey
| | | | - Fatih Oltulu
- Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey
| | - Huseyin Aktug
- Department of Histology and Embryology, Ege University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
43
|
Guan C, Zhang J, Zhang J, Shi H, Ni R. Enhanced expression of early mitotic inhibitor-1 predicts a poor prognosis in esophageal squamous cell carcinoma patients. Oncol Lett 2016; 12:114-120. [PMID: 27347110 PMCID: PMC4906579 DOI: 10.3892/ol.2016.4611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/08/2016] [Indexed: 01/15/2023] Open
Abstract
Early mitotic inhibitor-1 (Emi1), as a key cell cycle regulatory gene, induces S phase and mitotic entry by controlling anaphase-promoting complex substrates. Emi1 overexpression may be a prognostic factor for patients with invasive breast cancer. However, its expression and clinical significance in esophageal squamous cell carcinoma (ESCC) remain unknown. In the present study, Emi1 was overexpressed in ESCC samples, contrarily to their neighboring normal tissues. The expression of Emi1 was correlated with histological differentiation (P=0.032), lymphatic metastasis (P=0.006) and Ki-67 expression (P=0.028). Multivariate analysis indicated that the presence of lymphatic metastasis and the protein expression levels of Emi1 and Ki-67 were all independent prognostic factors for ESCC patients (P=0.042, 0.018 and 0.001, respectively). In vitro, however, the expression of Emi1 was upregulated in the ECA109 cell line following release from serum starvation. In addition, depletion of endogenous Emi1 by small interfering RNA could effectively reduce cell proliferation. Thus, the present data indicated that Emi1 expression was upregulated in ESCC tissues and correlated with poor survival in ESCC patients, and suggested that Emi1 may be an independent prognostic factor for ESCC patients.
Collapse
Affiliation(s)
- Chengqi Guan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianfeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hui Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
44
|
Bendris N, Lemmers B, Blanchard JM. Cell cycle, cytoskeleton dynamics and beyond: the many functions of cyclins and CDK inhibitors. Cell Cycle 2016; 14:1786-98. [PMID: 25789852 DOI: 10.1080/15384101.2014.998085] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
While targeting experiments carried out on the genes encoding many cell cycle regulators have challenged our views of cell cycle control, they also suggest that redundancy might not be the only explanation for the observed perplexing phenotypes. Indeed, several observations hint at functions of cyclins and CDK inhibitors that cannot be accounted for by their sole role as kinase regulators. They are found involved in many cellular transactions, depending or not on CDKs that are not directly linked to cell cycle control, but participating to general mechanisms such as transcription, DNA repair or cytoskeleton dynamics. In this review we discuss the roles that these alternative functions might have in cancer cell proliferation and migration that sometime even challenge their definition as proliferation markers.
Collapse
Affiliation(s)
- Nawal Bendris
- a Institut de Génétique Moléculaire de Montpellier; CNRS; Montpellier; France; Université Montpellier 2 ; Place Eugène Bataillon; Montpellier , France
| | | | | |
Collapse
|
45
|
Li W, Dong Q, Li L, Zhang Z, Cai X, Pan X. Prognostic significance of claudin-1 and cyclin B1 protein expression in patients with hypopharyngeal squamous cell carcinoma. Oncol Lett 2016; 11:2995-3002. [PMID: 27123052 PMCID: PMC4840523 DOI: 10.3892/ol.2016.4333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/19/2016] [Indexed: 11/25/2022] Open
Abstract
Claudin-l and cyclin B1 are abnormally expressed in certain malignancies, but their expression in hypopharyngeal squamous cell carcinoma (HSCC) has not been reported thus far. Studying the expression levels of claudin-1 and cylin B1 in HSCC tissues and their association with clinical stage, pathological grade and prognosis in patients with HSCC may provide a theoretical basis and guide future research on HSCC targeted therapy. The protein expression levels of the above two biomarkers was immunohistochemically detected in 97 HSCC cases and 90 matched adjacent tissue samples. The correlation between the expression levels of claudin-1 and cylin B1 and the patients' clinical parameters was analyzed via Pearson's χ2 test, while survival analysis was performed using a log-rank test. The results of the current study revealed that claudin-1 and cyclin B1 were highly expressed in HSCC tissues, and the expression of claudin-1 was associated with tumor differentiation degree and lymph node metastasis, while cyclin B1 expression was associated with tumor differentiation degree. Furthermore, Kaplan-Meier analysis revealed that claudin-1 expression correlated with survival (P=0.003), and the expression levels of claudin-1 and cyclin B1 were observed to be positively correlated, in patients with HSCC. Cyclin B1 and claudin-1 exhibited an elevated expression in HSCC specimens, thus suggesting their use as tumor markers. Therefore, the joint detection of claudin-1 and cyclin B1 may aid to guide cancer therapy and to determine prognosis in HSCC. Furthermore, claudin-1 may be used as an HSCC-monitoring index, and may serve as a therapeutic target.
Collapse
Affiliation(s)
- Wujie Li
- Department of Otolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China; Department of Otolaryngology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Qing Dong
- Department of Pediatrics, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Lei Li
- Department of Gastroenterology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Zhenlei Zhang
- Department of Otolaryngology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xiaolan Cai
- Department of Otolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xinliang Pan
- Department of Otolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
46
|
Buchegger K, Ili C, Riquelme I, Letelier P, Corvalán AH, Brebi P, Huang THM, Roa JC. Reprimo as a modulator of cell migration and invasion in the MDA-MB-231 breast cancer cell line. Biol Res 2016; 49:5. [PMID: 26796959 PMCID: PMC4722741 DOI: 10.1186/s40659-016-0066-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/08/2016] [Indexed: 12/21/2022] Open
Abstract
Background Reprimo (RPRM), a highly glycosylated protein, is a new downstream effector of p53-induced cell cycle arrest at the G2/M checkpoint, and a putative tumor suppressor gene frequently silenced via methylation of its promoter region in several malignances. The aim of this study was to characterize the epigenetic inactivation and its biological function in BC cell lines. Methods The correlation between RPRM methylation and loss of mRNA expression was assessed in six breast cancer cell lines by methylation specific PCR (MSP), 5′-Aza-2′-deoxycytidine treatment and RT-PCR assays. MDA-MB-231 cells were chosen to investigate the phenotypic effect of RPRM in cell proliferation, cell cycle, cell death, cell migration and invasion. Results In the cancer methylome system (CMS) (web-based system for visualizing and analyzing genome-wide methylation data of human cancers), the CpG island region of RPRM (1.1 kb) was hypermethylated in breast cancer compared to normal breast tissue; more interesting still was that ERα(+) tumors showed higher methylation intensity than ERα(−). Downregulation of RPRM mRNA by methylation was confirmed in MDA-MB-231 and BT-20 cell lines. In addition, overexpression of RPRM in MDA-MB-231 cells resulted in decreased rates of cell migration, wound healing and invasion in vitro. However, RPRM overexpression did not alter cell viability, phosphatidylserine (PS) translocation or G2/M cell cycle transition. Conclusion Taken together, these data suggest that RPRM is involved in decreased cell migration and invasion in vitro, acting as a potential tumor suppressor gene in the MDA-MB-231 cell line.
Collapse
Affiliation(s)
- Kurt Buchegger
- Department of Pathology, Molecular Pathology Laboratory BIOREN-CEGIN, School of Medicine, Universidad de La Frontera, Temuco, Chile.
| | - Carmen Ili
- Department of Pathology, Molecular Pathology Laboratory BIOREN-CEGIN, School of Medicine, Universidad de La Frontera, Temuco, Chile.
| | - Ismael Riquelme
- Department of Pathology, Molecular Pathology Laboratory BIOREN-CEGIN, School of Medicine, Universidad de La Frontera, Temuco, Chile.
| | - Pablo Letelier
- School of Health Sciences, Universidad Católica de Temuco, Temuco, Chile.
| | - Alejandro H Corvalán
- Centre for Translational Research in Oncology (CITO) and Department of Hematology and Oncology, Pontificia Universidad Catolica de Chile, Santiago, Chile.
| | - Priscilla Brebi
- Department of Pathology, Molecular Pathology Laboratory BIOREN-CEGIN, School of Medicine, Universidad de La Frontera, Temuco, Chile.
| | - Tim Hui-Ming Huang
- Department of Molecular Medicine/Institute of Biotechnology, University of Texas Health Science Center at San Antonio, STRF, Room 225, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| | - Juan Carlos Roa
- Department of Pathology, Advanced Center for Chronic Diseases (ACCDiS) (CITO), School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
47
|
Kaddi CD, Wang MD. Models for Predicting Stage in Head and Neck Squamous Cell Carcinoma Using Proteomic and Transcriptomic Data. IEEE J Biomed Health Inform 2015; 21:246-253. [PMID: 26462248 DOI: 10.1109/jbhi.2015.2489158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Late diagnosis is one of the reasons that head and neck squamous cell carcinoma (HNSCC) patients experience relative five-year survival rates ranging from 40%-66%. The molecular-level differences between early and advanced stage HNSCC may provide insight into therapeutic targets and strategies. Previous bioinformatics studies have shown mixed or limited results in identifying gene and protein markers and in developing models for discriminating between early and advanced stage HNSCC. Thus, we have investigated models for HNSCC stage prediction using RNAseq and reverse phase protein array data from The Cancer Genome Atlas and The Cancer Proteome Atlas. We systematically assessed individual and ensemble binary classifiers, using filter and wrapper feature selection methods, to develop several well-performing models. In particular, integrated models harnessing both data types consistently resulted in better performance. This study identifies informative protein and gene feature sets which may increase understanding of HNSCC progression.
Collapse
|
48
|
Shang ZF, Tan W, Liu XD, Yu L, Li B, Li M, Song M, Wang Y, Xiao BB, Zhong CG, Guan H, Zhou PK. DNA-PKcs Negatively Regulates Cyclin B1 Protein Stability through Facilitating Its Ubiquitination Mediated by Cdh1-APC/C Pathway. Int J Biol Sci 2015. [PMID: 26221070 PMCID: PMC4515814 DOI: 10.7150/ijbs.12443] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) is a critical component of the non-homologous end-joining pathway of DNA double-stranded break repair. DNA-PKcs has also been shown recently functioning in mitotic regulation. Here, we report that DNA-PKcs negatively regulates the stability of Cyclin B1 protein through facilitating its ubiquitination mediated by Cdh1 / E 3 ubiquitin ligase APC/C pathway. Loss of DNA-PKcs causes abnormal accumulation of Cyclin B1 protein. Cyclin B1 degradation is delayed in DNA-PKcs-deficient cells as result of attenuated ubiquitination. The impact of DNA-PKcs on Cyclin B1 stability relies on its kinase activity. Our study further reveals that DNA-PKcs interacts with APC/C core component APC2 and its co-activator Cdh1. The destruction of Cdh1 is accelerated in the absence of DNA-PKcs. Moreover, overexpression of exogenous Cdh1 can reverse the increase of Cyclin B1 protein in DNA-PKcs-deficient cells. Thus, DNA-PKcs, in addition to its direct role in DNA damage repair, functions in mitotic progression at least partially through regulating the stability of Cyclin B1 protein.
Collapse
Affiliation(s)
- Zeng-Fu Shang
- 1. School of Radiation Medicine and Protection, Medical College of Soochow University; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu 215123, P. R. China ; 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Wei Tan
- 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China ; 3. School of Public Heath, Central South University, Changsha, Hunan Province, Changsha, Hunan 410078, P. R. China
| | - Xiao-Dan Liu
- 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Lan Yu
- 4. Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, USA
| | - Bing Li
- 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Ming Li
- 1. School of Radiation Medicine and Protection, Medical College of Soochow University; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu 215123, P. R. China
| | - Man Song
- 1. School of Radiation Medicine and Protection, Medical College of Soochow University; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu 215123, P. R. China ; 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Yu Wang
- 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Bei-Bei Xiao
- 1. School of Radiation Medicine and Protection, Medical College of Soochow University; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu 215123, P. R. China
| | - Cai-Gao Zhong
- 3. School of Public Heath, Central South University, Changsha, Hunan Province, Changsha, Hunan 410078, P. R. China
| | - Hua Guan
- 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| | - Ping-Kun Zhou
- 1. School of Radiation Medicine and Protection, Medical College of Soochow University; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu 215123, P. R. China ; 2. Department of Radiation Toxicology and Oncology, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing Institute of Radiation Medicine, Beijing 100850, P. R. China
| |
Collapse
|
49
|
Fang Y, Liang X, Jiang W, Li J, Xu J, Cai X. Cyclin b1 suppresses colorectal cancer invasion and metastasis by regulating e-cadherin. PLoS One 2015; 10:e0126875. [PMID: 25962181 PMCID: PMC4427130 DOI: 10.1371/journal.pone.0126875] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 04/08/2015] [Indexed: 02/06/2023] Open
Abstract
Cyclin B1, a mitotic cyclin, has been implicated in malignances. However, its contribution to colorectal cancer invasion and metastasis are still not well understood. Here, we demonstrated that the invasion and metastasis of colorectal cancer is regulated by Cyclin B1. Overexpression of Cyclin B1 was observed in colorectal cancer tissues, but this elevated expression was negatively associated with lymph node metastasis, distant metastasis stage, and TNM stage. The Kaplan-Meier survival analysis proved that low Cyclin B1 expression was associated with poor overall survival of patients with colorectal cancer. Inhibition of Cyclin B1 in colorectal cancer cells enhanced the cell migration and invasion of three different colorectal cancer cell lines. In studying the possible mechanism by which Cyclin B1 suppresses colorectal cancer invasion and metastasis, we observed that suppression of Cyclin B1 decreased the expression of E-cadherin protein level. Our findings suggest that Cyclin B1 could suppress the invasion and metastasis of colorectal cancer cells through regulating E-cadherin expression, which enables the development of potential intervention strategies for colorectal cancer.
Collapse
Affiliation(s)
- Yifeng Fang
- The Second Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Xiao Liang
- The Second Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Wenbin Jiang
- The Second Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Jianbo Li
- The Second Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xiujun Cai
- The Second Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- * E-mail:
| |
Collapse
|
50
|
CCNB1 is a prognostic biomarker for ER+ breast cancer. Med Hypotheses 2014; 83:359-64. [DOI: 10.1016/j.mehy.2014.06.013] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/08/2014] [Accepted: 06/15/2014] [Indexed: 12/15/2022]
|