1
|
Lei X, Qiu L, Chen Q, Liao L, Yu P, Wu W, Zhu Z, Li C, Lin G, Zhuang Z, Meng Y, Wang Y, Wang C, Du Y. Exploring the regulatory mechanism of CCNA2 in colorectal cancer: insights from multiomics and experimental analysis. J Biol Chem 2025:110216. [PMID: 40345591 DOI: 10.1016/j.jbc.2025.110216] [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: 05/11/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
Colorectal cancer (CRC) is the third-most common cancer and the second-leading cause of mortality due to cancer worldwide. The underlying regulatory mechanism of CCNA2 in CRC was explored through multiomics and experimental analyses, thus facilitating diagnosis, therapy and prognosis. Two gene expression datasets (i.e., GSE9348 and GSE110223) were extracted from GEO. Differentially expressed genes (DEGs) were identified via GEO2R, which were used for enrichment analyses through DAVID. PPI network of DEGs was constructed by STRING, and the core genes were identified. CCNA2, a prognostic core gene for CRC, was validated in TCGA and HPA via transcriptomics and proteomics. ROC analysis was performed to evaluate the diagnostic value of CCNA2 in CRC. The therapeutic value of CCNA2 was evaluated in DGIdb through pharmacogenomics. The correlation between CCNA2 and immune infiltration was determined in TIMER by immunomics. TF-mRNA and miRNA-mRNA networks for CCNA2 were constructed in miRnet and miRDB via transcriptomics. The role and mechanism of CCNA2 in CRC were investigated both in vitro and in vivo. The miR-548x-3p/CCNA2 regulatory axis in CRC was investigated in vitro. CCNA2 showed excellent diagnostic, therapeutic, and prognostic value in CRC. CCNA2 was closely associated with tumor-infiltrating immunocytes, TFs, and miRNAs. The upregulation of CCNA2 was observed in CRC, and the knockdown of CCNA2 inhibited the proliferation, migration, and invasion while inducing apoptosis of CRC cells. The knockdown of CCNA2 could inhibit epithelial-mesenchymal transition (EMT) pathway. CCNA2 acted as a target of miR-548x-3p in regulating the biological behavior of CRC cells via the EMT-signaling pathway. CCNA2 is a potential biomarker for the diagnosis, treatment, and prognosis of CRC and is associated with immune infiltration, TF, and miRNA. The miR-548x-3p/CCNA2 axis plays a pivotal role in regulating the tumorigenesis of CRC through the EMT-signaling pathway.
Collapse
Affiliation(s)
- Xinyi Lei
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Lanying Qiu
- Department of Chest Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Qiang Chen
- Department of Oncology, Cancer Diagnosis and Therapy Research Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Lan Liao
- Department of Pathology, the First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
| | - Pengfei Yu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wenjie Wu
- Department of Radiation Physics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhengyang Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chunying Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Gang Lin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, Zhejiang 310024, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zirui Zhuang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, Zhejiang 310024, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuxin Meng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yan Wang
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Cunchuan Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China.
| | - Yian Du
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
2
|
Zhao X, Ha M, Zhou L, Wang Y, Li P. Berberine diminishes the malignant progression of non-small cell lung cancer cells by targeting CDCA5 and CCNA2. J Nat Med 2025; 79:530-542. [PMID: 40155519 DOI: 10.1007/s11418-025-01885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/13/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Berberine (BBR), an isoquinoline alkaloid from Coptidis Rhizoma, possesses powerful activities against diverse human malignancies, including non-small cell lung cancer (NSCLC). Nevertheless, the underlying anti-tumor mechanisms of BBR in NSCLC remain poorly understood. METHODS NSCLC cells were cultured and treated with various doses (0, 15, 30, and 45 μM) of BBR for 48 h. Cell viability, proliferation, apoptosis, migration, and invasion were detected using 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell, and wound healing assays. Cell division cycle-associated protein 5 (CDCA5) and Cyclin A2 (CCNA2) mRNA level and protein level were measured using real-time quantitative polymerase chain reaction (RT-qPCR) and western blot assays. After STRING databases prediction, the possible interaction between CDCA5 and CCNA2 was identified using Co-Immunoprecipitation (IP) assays. The biological role of BBR treatment on NSCLC tumor growth was assessed using the xenograft tumor model in vivo. RESULTS BBR treatment blocked NSCLC cell proliferation, migration, invasion, and promoted apoptosis. CDCA5 and CCNA2 levels were increased in NSCLC tissues, whereas their expression was decreased in BBR-induced NSCLC cells. CDCA5 or CCNA2 overexpression might attenuate the inhibitory role of BBR on NSCLC cell malignant behaviors. CDCA5 interacted with CCNA2 to regulate its expression in NSCLC cells. BBR administration blocked NSCLC xenograft growth in vivo. CONCLUSION BBR hindered NSCLC cell malignant progression partly by modulating CDCA5 and CCNA2, providing a promising therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5, Renmin Street, Guta District, Jinzhou City, 121000, Liaoning Province, China
| | - Minwen Ha
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5, Renmin Street, Guta District, Jinzhou City, 121000, Liaoning Province, China
| | - Lulu Zhou
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5, Renmin Street, Guta District, Jinzhou City, 121000, Liaoning Province, China
| | - Yanyun Wang
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5, Renmin Street, Guta District, Jinzhou City, 121000, Liaoning Province, China
| | - Ping Li
- Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, No.2, Section 5, Renmin Street, Guta District, Jinzhou City, 121000, Liaoning Province, China.
| |
Collapse
|
3
|
Hoang HNQ, Ho CNQ, Dang LTT, Phan NLC, Doan CC, Nguyen HTM, Le CPM, Hoang SN, Le LT. The Proliferation of Chang Liver Cells After Simulated Microgravity Induction. Curr Issues Mol Biol 2025; 47:164. [PMID: 40136418 PMCID: PMC11941360 DOI: 10.3390/cimb47030164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
This study aimed to assess the recovery capability of Chang liver cells (CCL-13) following simulated microgravity (SMG) induction. CCL-13 cells were cultured under SMG conditions for 72 h, and control group cells were cultured under 1G conditions for an identical duration. Cells from the SMG and control groups were further cultured under 1G conditions and assessed after 24 h and 72 h intervals in the gravity recovery experiment. The WST1 results indicated that CCL-13 proliferation was more evident in the control group than in the SMG group after both the 24 h and 72 h intervals. The control group had a lower percentage of CCL-13 cells in the G0/G1 phase compared with the SMG group at both time points, and it exhibited a higher total percentage of cells in the S and G2/M phases. The control group exhibited elevated levels of cell-cycle-related proteins, including cyclin A, cyclin D, and cdk6, compared with the SMG group. The flow cytometry results revealed that the apoptotic rate in the control group was significantly lower than that in the SMG group at both the 24 h and 72 h time points. However, the apoptotic percentage in the SMG group at the 72-h mark was significantly lower than that at the 24-h mark. SMG reduces the viability and proliferation ability of CCL-13 cells. After a period of recovery and adaptation to normal gravity conditions (1G), the CCL-13 cells in the SMG group showed better signs of recovery after 72 h than after 24 h.
Collapse
Affiliation(s)
- Huy Nghia Quang Hoang
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
- Biotechnology Department, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi 100000, Vietnam
| | - Chi Nguyen Quynh Ho
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
| | - Loan Thi Tung Dang
- Faculty of Biology and Biotechnology, University of Science, Vietnam National University, Ho Chi Minh 700000, Vietnam;
| | - Nhan Lu Chinh Phan
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
| | - Chung Chinh Doan
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
- Biotechnology Department, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi 100000, Vietnam
| | - Han Thai Minh Nguyen
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Life Science Department, University of New Hampshire at Manchester, Manchester, NH 03101, USA
| | - Cuong Phan Minh Le
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
| | - Son Nghia Hoang
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
- Biotechnology Department, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi 100000, Vietnam
| | - Long Thanh Le
- Animal Biotechnology Department, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh 700000, Vietnam; (H.N.Q.H.); (C.N.Q.H.); (N.L.C.P.); (C.C.D.); (H.T.M.N.); (C.P.M.L.); (S.N.H.)
- Biotechnology Department, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi 100000, Vietnam
| |
Collapse
|
4
|
Almier N, Leibowitz K, Gower AC, To S, Keller MR, Connizzo BK, Roh DS, Alani RM, Collard M. Targeting the Epigenome Reduces Keloid Fibroblast Cell Proliferation, Migration, and Invasion. J Invest Dermatol 2025; 145:411-422.e7. [PMID: 39009280 PMCID: PMC11995714 DOI: 10.1016/j.jid.2024.06.1274] [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: 07/18/2023] [Revised: 05/16/2024] [Accepted: 06/03/2024] [Indexed: 07/17/2024]
Abstract
Keloids are pathological fibroproliferative scars resulting from abnormal collagen deposition within and beyond the margins of the initial cutaneous insult. Keloids negatively impact QOL functionally and cosmetically, with current treatment modalities unsatisfactory. Recent studies indicate that epigenetic dysregulation is central to the development and progression of keloids. In this study, we evaluate the functional significance of epigenetic targeting strategies in vitro using patient-derived keloid fibroblasts treated with small-molecule inhibitors of histone deacetylases, LSD1, CoREST, and p300, as potential therapies for keloids. We find that both the dual-acting CoREST inhibitor corin and the histone deacetylase inhibitor entinostat reduce fibroblast proliferation more than the LSD1 inhibitor GSK-LSD1; in addition, corin was the most effective inhibitor of migration and invasion across keloid fibroblasts. RNA-sequencing analysis of keloid fibroblasts treated with corin demonstrates coordinate upregulation of many genes, including key mediators of cell adhesion such as claudins. Corin also downregulates gene sets involved in cell cycle progression, including reduced expression of cyclins A1 and B2 compared with that of DMSO. These results highlight a significant role for epigenetic regulation of pathologic mediators of keloidal scarring and suggest that inhibitors of the epigenetic CoREST repressor complex may prove beneficial in the prevention and/or treatment of keloidal scarring in patients.
Collapse
Affiliation(s)
- Nedaa Almier
- Department of Dermatology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Kassidy Leibowitz
- Department of Dermatology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Samantha To
- Department of Dermatology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Madelyn R Keller
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, Massachusetts, USA
| | - Brianne K Connizzo
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, Massachusetts, USA
| | - Daniel S Roh
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Rhoda M Alani
- Department of Dermatology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA.
| | - Marianne Collard
- Department of Dermatology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
5
|
Chen Z, Zeng Y, Ma P, Xu Q, Zeng L, Song X, Yu F. Integrated GMPS and RAMP3 as a signature to predict prognosis and immune heterogeneity in hepatocellular carcinoma. Gene 2025; 933:148958. [PMID: 39312983 DOI: 10.1016/j.gene.2024.148958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/14/2024] [Accepted: 09/18/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly fatal malignant worldwide. As different expression levels of specific genes can lead to different HCC outcomes, we aimed to develop a gene signature capable of predicting HCC prognosis. METHODS In this study, transcriptomic sequencing and relevant clinical data were extracted from public platforms. The guanine monophosphate synthase (GMPS)|receptor activity-modifying protein 3 (RAMP3) gene pair was developed based on the relative values of gene expression levels. Nomograms were developed using R software. Immune status was assessed through single-sample gene set enrichment analysis. GMPS knockdown was achieved through siRNA transfection. Quantitative reverse transcription PCR, apoptosis assays, and cell proliferation were performed to verify the function of GMPS|RAMP3 in HCC cells. RESULTS Here, a gene pair containing GMPS and RAMP3 was successfully constructed. We demonstrated that the GMPS|RAMP3 gene pair was an independent predictor with strong prognostic prediction power, based on which a nomogram was established. Functional analysis revealed that the enrichment of cell cycle-related pathways and immune status differed considerably between the two groups, with cell cycle-related genes highly expressed in the high GMPS|RAMP3 value group. Finally, cell experiments indicated that GMPS knockdown significantly repressed proliferation, promoted apoptosis, and enhanced the sensitivity of HCC cells to gemcitabine. CONCLUSIONS The gene pair GMPS|RAMP3 is a novel prognostic predictor of HCC, providing a promising approach to the treatment and assessment of immune heterogeneity in HCC.
Collapse
Affiliation(s)
- Zhuoyan Chen
- Department of Gastroenterology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Yuan Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peipei Ma
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qian Xu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liuwei Zeng
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xian Song
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fujun Yu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
6
|
Kubat Oktem E, Demir U, Yazar M, Arga KY. Three candidate anticancer drugs were repositioned by integrative analysis of the transcriptomes of species with different regenerative abilities after injury. Comput Biol Chem 2023; 106:107934. [PMID: 37487250 DOI: 10.1016/j.compbiolchem.2023.107934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
Regeneration is a homeostatic process that involves the restoration of cells and body parts. Most of the molecular mechanisms and signalling pathways involved in wound healing, such as proliferation, have also been associated with cancer cell growth, suggesting that cancer is an over/unhealed wound. In this study, we examined differentially expressed genes in spinal cord samples from regenerative organisms (axolotl and zebrafish) and nonregenerative organisms (mouse and rat) compared to intact control spinal cord samples using publicly available transcriptomics data and bioinformatics analyses. Based on these gene signatures, we investigated 3 small compounds, namely cucurbitacin I, BMS-754807, and PHA-793887 as potential candidates for the treatment of cancer. The predicted target genes of the repositioned compounds were mainly enriched with the greatest number of genes in cancer pathways. The molecular docking results on the binding affinity between the repositioned compounds and their target genes are also reported. The repositioned 3 small compounds showed anticancer effect both in 2D and 3D cell cultures using the prostate cancer cell line as a model. We propose cucurbitacin I, BMS-754807, and PHA-793887 as potential anticancer drug candidates. Future studies on the mechanisms associated with the revealed gene signatures and anticancer effects of these three small compunds would allow scientists to develop therapeutic approaches to combat cancer. This research contributes to the evaluation of mechanisms and gene signatures that either limit or cause cancer, and to the development of new cancer therapies by establishing a link between regeneration and carcinogenesis.
Collapse
Affiliation(s)
- Elif Kubat Oktem
- Department of Molecular Biology and Genetics, Istanbul Medeniyet University, Istanbul, Turkey.
| | - Ummuhan Demir
- Department of Molecular Biology and Genetics, Istanbul Medeniyet University, Istanbul, Turkey; Istanbul Medeniyet University, Science and Advanced Technology Research Center (BILTAM), Istanbul, Turkey
| | - Metin Yazar
- Department of Genetics and Bioengineering, Istanbul Okan University, Istanbul, Turkey; Department of Bioengineering, Marmara University, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, Istanbul, Turkey; Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, Istanbul, Turkey
| |
Collapse
|
7
|
Guan Y, Sun Y, Liu Z, Zhang Y, Cao M, Wang W, Tao J, Yao Y. INSM1 promotes breast carcinogenesis by regulating C-MYC. Am J Cancer Res 2023; 13:3500-3516. [PMID: 37693125 PMCID: PMC10492136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/28/2023] [Indexed: 09/12/2023] Open
Abstract
Insulinoma-associated protein-1 (INSM1), which is highly expressed in various neuroendocrine tumors, functions as a zinc finger transcription factor capable of regulating the biological behavior of tumor cells. However, its specific role in breast cancer remains unclear. This study aims to investigate the role and mechanism of INSM1 in breast cancer. A total of 158 cohorts were recruited to examine the expression of INSM1 in breast cancer tissues and their corresponding adjacent normal tissues using immunohistochemistry. Follow-up data, along with clinical and pathological information, were collected to analyze the correlation between INSM1 expression and survival outcomes in breast cancer patients. Additionally, we investigated the impact of INSM1 on breast cancer cell proliferation, migration, and aggregation. To further explore the regulatory effect of INSM1 knockdown on breast cancer tumor growth, we utilized a xenograft mouse model. The results revealed that INSM1 was significantly overexpressed in breast cancer patients and correlated with prognosis. Knockdown of INSM1 notably impaired the malignant biological effects of breast cancer cells and inhibited the growth of xenograft tumors in nude mice. Importantly, our data also suggests an interaction between INSM1 and S-phase kinase-associated protein 2 (SKP2), which in turn regulates C-MYC, thereby affecting the p-ERK pathway. Our study provides the first evidence demonstrating the contribution of INSM1 to tumor formation and growth in breast cancer. Furthermore, we found that INSM1 positively regulates C-MYC and the p-ERK pathway by interacting with SKP2 during breast cancer development. Collectively, these findings highlight INSM1 as a promising target for breast cancer treatment.
Collapse
Affiliation(s)
- Yinan Guan
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| | - Yulu Sun
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| | - Zheying Liu
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
- Department of Clinical Medicine, Southeast Univeristy SchoolNanjing 210008, Jiangsu, China
| | - Yin Zhang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| | - Meng Cao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| | - Wei Wang
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| | - Jinqiu Tao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| | - Yongzhong Yao
- Division of Breast Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing UniversityNanjing 210008, Jiangsu, China
| |
Collapse
|
8
|
Elshaer SS, Abulsoud AI, Fathi D, Abdelmaksoud NM, Zaki MB, El-Mahdy HA, Ismail A, Elsakka EGE, Abd-Elmawla MA, Abulsoud LA, Doghish AS. miRNAs role in glioblastoma pathogenesis and targeted therapy: Signaling pathways interplay. Pathol Res Pract 2023; 246:154511. [PMID: 37178618 DOI: 10.1016/j.prp.2023.154511] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
High mortality and morbidity rates and variable clinical behavior are hallmarks of glioblastoma (GBM), the most common and aggressive primary malignant brain tumor. Patients with GBM often have a dismal outlook, even after undergoing surgery, postoperative radiation, and chemotherapy, which has fueled the search for specific targets to provide new insights into the development of contemporary therapies. The ability of microRNAs (miRNAs/miRs) to posttranscriptionally regulate the expression of various genes and silence many target genes involved in cell proliferation, cell cycle, apoptosis, invasion, angiogenesis, stem cell behavior and chemo- and radiotherapy resistance makes them promising candidates as prognostic biomarkers and therapeutic targets or factors to advance GBM therapeutics. Hence, this review is like a crash course in GBM and how miRNAs related to GBM. Here, we will outline the miRNAs whose role in the development of GBM has been established by recent in vitro or in vivo research. Moreover, we will provide a summary of the state of knowledge regarding oncomiRs and tumor suppressor (TS) miRNAs in relation to GBM with an emphasis on their potential applications as prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Shereen Saeid Elshaer
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo 11823, Egypt; Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed I Abulsoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Doaa Fathi
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Logyna A Abulsoud
- Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
9
|
Huffman KE, Li LS, Carstens R, Park H, Girard L, Avila K, Wei S, Kollipara R, Timmons B, Sudderth J, Bendris N, Kim J, Villalobos P, Fujimoto J, Schmid S, Deberardinis RJ, Wistuba I, Heymach J, Kittler R, Akbay EA, Posner B, Wang Y, Lam S, Kliewer SA, Mangelsdorf DJ, Minna JD. Glucocorticoid mediated inhibition of LKB1 mutant non-small cell lung cancers. Front Oncol 2023; 13:1025443. [PMID: 37035141 PMCID: PMC10078807 DOI: 10.3389/fonc.2023.1025443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/20/2023] [Indexed: 04/11/2023] Open
Abstract
The glucocorticoid receptor (GR) is an important anti-cancer target in lymphoid cancers but has been understudied in solid tumors like lung cancer, although glucocorticoids are often given with chemotherapy regimens to mitigate side effects. Here, we identify a dexamethasone-GR mediated anti-cancer response in a subset of aggressive non-small cell lung cancers (NSCLCs) that harbor Serine/Threonine Kinase 11 (STK11/LKB1) mutations. High tumor expression of carbamoyl phosphate synthase 1 (CPS1) was strongly linked to the presence of LKB1 mutations, was the best predictor of NSCLC dexamethasone (DEX) sensitivity (p < 10-16) but was not mechanistically involved in DEX sensitivity. Subcutaneous, orthotopic and metastatic NSCLC xenografts, biomarker-selected, STK11/LKB1 mutant patient derived xenografts, and genetically engineered mouse models with KRAS/LKB1 mutant lung adenocarcinomas all showed marked in vivo anti-tumor responses with the glucocorticoid dexamethasone as a single agent or in combination with cisplatin. Mechanistically, GR activation triggers G1/S cell cycle arrest in LKB1 mutant NSCLCs by inducing the expression of the cyclin-dependent kinase inhibitor, CDKN1C/p57(Kip2). All findings were confirmed with functional genomic experiments including CRISPR knockouts and exogenous expression. Importantly, DEX-GR mediated cell cycle arrest did not interfere with NSCLC radiotherapy, or platinum response in vitro or with platinum response in vivo. While DEX induced LKB1 mutant NSCLCs in vitro exhibit markers of cellular senescence and demonstrate impaired migration, in vivo DEX treatment of a patient derived xenograft (PDX) STK11/LKB1 mutant model resulted in expression of apoptosis markers. These findings identify a previously unknown GR mediated therapeutic vulnerability in STK11/LKB1 mutant NSCLCs caused by induction of p57(Kip2) expression with both STK11 mutation and high expression of CPS1 as precision medicine biomarkers of this vulnerability.
Collapse
Affiliation(s)
- Kenneth E. Huffman
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Long Shan Li
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ryan Carstens
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hyunsil Park
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Luc Girard
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kimberley Avila
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Shuguang Wei
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Rahul Kollipara
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Brenda Timmons
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jessica Sudderth
- Children’s Medical Center Research Institute at University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| | - Nawal Bendris
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jiyeon Kim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
- Department of Urology, Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, United States
| | - Pamela Villalobos
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sandra Schmid
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ralph J. Deberardinis
- Children’s Medical Center Research Institute at University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Ralf Kittler
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Esra A. Akbay
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yuzhuo Wang
- British Columbia Cancer Center, Vancouver, BC, Canada
| | - Stephen Lam
- British Columbia Cancer Center, Vancouver, BC, Canada
| | - Steven A. Kliewer
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David J. Mangelsdorf
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John D. Minna
- Department of Internal Medicine, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
10
|
Miller KJ, Henry I, Maylin Z, Smith C, Arunachalam E, Pandha H, Asim M. A compendium of Androgen Receptor Variant 7 target genes and their role in Castration Resistant Prostate Cancer. Front Oncol 2023; 13:1129140. [PMID: 36937454 PMCID: PMC10014620 DOI: 10.3389/fonc.2023.1129140] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Persistent androgen receptor (AR) signalling is the main driver of prostate cancer (PCa). Truncated isoforms of the AR called androgen receptor variants (AR-Vs) lacking the ligand binding domain often emerge during treatment resistance against AR pathway inhibitors such as Enzalutamide. This review discusses how AR-Vs drive a more aggressive form of PCa through the regulation of some of their target genes involved in oncogenic pathways, enabling disease progression. There is a pressing need for the development of a new generation of AR inhibitors which can repress the activity of both the full-length AR and AR-Vs, for which the knowledge of differentially expressed target genes will allow evaluation of inhibition efficacy. This review provides a detailed account of the most common variant, AR-V7, the AR-V7 regulated genes which have been experimentally validated, endeavours to understand their relevance in aggressive AR-V driven PCa and discusses the utility of the downstream protein products as potential drug targets for PCa treatment.
Collapse
Affiliation(s)
| | | | - Zoe Maylin
- *Correspondence: Zoe Maylin, ; Mohammad Asim,
| | | | | | | | | |
Collapse
|
11
|
Yang X, Liu Y, Zhou X, Chen K, Xu J, Xu S. Circular RNA 0010117 promotes aggressive glioblastoma behavior by regulating the miRNA-6779-5p/SPEN axis. Transl Oncol 2022; 25:101515. [PMID: 36087384 PMCID: PMC9468456 DOI: 10.1016/j.tranon.2022.101515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 01/16/2023] Open
Abstract
Circ-0010117 is down-regulated in glioblastoma. Circ-0010117 regulates aggressiveness via miRNA-6779-5p in glioblastoma. SPEN contributed to regulate miRNA-6779-5p in glioblastoma. Upregulated Circ-0010117 inhibited in vivo tumor growth of human glioblastoma xenograft.
Noncoding RNAs (ncRNAs) play important roles in cancer biology, providing potential targets for cancer intervention. As a new class of endogenous noncoding RNAs, circular RNAs (circRNAs) have been recently identified in cell development and function, and certain types of pathological responses contribute to cancer progression, including glioblastoma. However, the potential mechanisms underlying the relationship between circRNAs and glioblastoma progression are still largely unknown. Methods: The expression and roles of circular RNA 0010117 (circ-0010117) were examined in vitro and in vivo. Quantitative RT‒PCR and western blotting were used to measure the expression of circRNA, miRNA, each gene, or related proteins. Cell biology experiments were performed to detect the biological function of circ-0010117 in glioblastoma cell lines. Moreover, bioinformatics analysis, luciferase reporter assays, and functional complementation analysis were carried out to investigate the target genes. Tumorigenesis was also evaluated by xenografting cells into nude mice. In this study, we found that circ-0010117 is downregulated in glioblastoma compared with corresponding paratumoural tissues. Subsequently, we observed that circ-0010117 can regulate aggressiveness in glioblastoma cells through miR-6779-5p. Furthermore, SPEN was verified as a direct target of miR-6779-5p and contributes to the circ-0010117 regulatory network. In addition, we identified that overexpression of circ-0010117 can suppress tumorigenesis in nude mice. Our findings indicate that circular RNA 0010117 promotes the aggressive behavior of glioblastoma by regulating the miRNA-6779-5p/SPEN axis. Our results provide a rationale for the use of circ-0010117 as a novel potential therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Xuanyong Yang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, China
| | - Yue Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, China
| | - Xinhui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, China; Institute of Medicine, Nanchang University, China
| | - Kang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, China
| | - Jiang Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, China.
| | - Shan Xu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, 17 Yong Wai Street, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
12
|
Dulińska-Litewka J, Felkle D, Dykas K, Handziuk Z, Krzysztofik M, Gąsiorkiewicz B. The role of cyclins in the development and progression of prostate cancer. Biomed Pharmacother 2022; 155:113742. [PMID: 36179490 DOI: 10.1016/j.biopha.2022.113742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/10/2022] [Accepted: 09/21/2022] [Indexed: 11/02/2022] Open
Abstract
The role of cyclins in hormone-dependent neoplasms is crucial in the development of the disease that is resistant to first-line therapy, as the example of breast cancer shows. However, in prostate cancer, cyclins are studied to a lesser extent. There are some well-described molecular pathways, including cyclins A1 and D1 signaling, however the role of other cyclins, e.g., D2, D3, E, and H, still requires further investigation. Recent studies indicate that cyclins regulate various cellular processes, not only the cell cycle. Furthermore, they remain in cross-talk with many other signaling pathways, e.g., MAPK/ERK, PI3K/Akt, and Notch. The androgen signaling axis, which is pivotal in prostate cancer progression, interferes with cyclin pathways at many levels. This article summarizes current knowledge on the influence of cyclins on prostate cancer progression by describing interactions between the androgen receptor and cyclins, as well as mechanisms underlying the development of resistance to currently used therapies.
Collapse
Affiliation(s)
- Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Mikołaja Kopernika Street 7C, Poland.
| | - Dominik Felkle
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Mikołaja Kopernika Street 7C, Poland
| | - Kacper Dykas
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Mikołaja Kopernika Street 7C, Poland
| | - Zuzanna Handziuk
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Mikołaja Kopernika Street 7C, Poland
| | - Marta Krzysztofik
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Mikołaja Kopernika Street 7C, Poland
| | - Bartosz Gąsiorkiewicz
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Krakow, Mikołaja Kopernika Street 7C, Poland
| |
Collapse
|
13
|
Kumari P, Kumar S, Sethy M, Bhue S, Mohanta BK, Dixit A. Identification of therapeutically potential targets and their ligands for the treatment of OSCC. Front Oncol 2022; 12:910494. [PMID: 36203433 PMCID: PMC9530560 DOI: 10.3389/fonc.2022.910494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Recent advancements in cancer biology have revealed molecular changes associated with carcinogenesis and chemotherapeutic exposure. The available information is being gainfully utilized to develop therapies targeting specific molecules involved in cancer cell growth, survival, and chemoresistance. Targeted therapies have dramatically increased overall survival (OS) in many cancers. Therefore, developing such targeted therapies against oral squamous cell carcinoma (OSCC) is anticipated to have significant clinical implications. In the current work, we have identified drug-specific sensitivity-related prognostic biomarkers (BOP1, CCNA2, CKS2, PLAU, and SERPINE1) using gene expression, Cox proportional hazards regression, and machine learning in OSCC. Dysregulation of these markers is significantly associated with OS in many cancers. Their elevated expression is related to cellular proliferation and aggressive malignancy in various cancers. Mechanistically, inhibition of these biomarkers should significantly reduce cellular proliferation and metastasis in OSCC and should result in better OS. It is pertinent to note that no effective small-molecule candidate has been identified against these biomarkers to date. Therefore, a comprehensive in silico drug design strategy assimilating homology modeling, extensive molecular dynamics (MD) simulation, and ensemble molecular docking has been applied to identify potential compounds against identified targets, and potential molecules have been identified. We hope that this study will help in deciphering potential genes having roles in chemoresistance and a significant impact on OS. It will also result in the identification of new targeted therapeutics against OSCC.
Collapse
Affiliation(s)
- Pratima Kumari
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Sugandh Kumar
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | - Madhusmita Sethy
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
| | - Shyamlal Bhue
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Bineet Kumar Mohanta
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Anshuman Dixit
- Computational Biology and Bioinformatics Laboratory, Institute of Life Sciences, Bhubaneswar, India
- *Correspondence: Anshuman Dixit,
| |
Collapse
|
14
|
Kusumastuti R, Kumagai Y, Ishihara S, Enomoto A, Murakami T, Yasuda M, Haga H. Mammaglobin 1 mediates progression of trastuzumab-resistant breast cancer cells through regulation of cyclins and NF-κB. FEBS Open Bio 2022; 12:1797-1813. [PMID: 35945910 PMCID: PMC9527592 DOI: 10.1002/2211-5463.13468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/15/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022] Open
Abstract
Overexpression of human epidermal growth factor receptor 2 (HER2) in various cancers is correlated with poor patient survival. Trastuzumab, a recombinant humanized monoclonal antibody against HER2, has been considered to be a first-line therapy for HER2-positive breast cancer patients, but its usefulness is limited by the development of resistance. In this study, we established resistant cells by long-term treatment with trastuzumab. These cells showed higher proliferation, invasion, and migration abilities than the wild-type cells. Mammaglobin 1 (MGB1), cyclin D1, E1, A2, and phosphorylated NF-κB (p-p65) were upregulated in resistant cells. These proteins regulate cell proliferation, migration, and invasion of resistant cells. Depletion of MGB1 decreased cyclin and p-p65 expression. Cyclin D1 and A2, but not E1 expression, were affected by p-p65 downregulation. In summary, our results indicate that MGB1 expression is increased in breast cancer cells that have gained resistance to trastuzumab, and suggest that MGB1 promotes aggressiveness through cyclin and NF-κB regulation.
Collapse
Affiliation(s)
- Ratih Kusumastuti
- Division of Life Science, Graduate School of Life ScienceHokkaido UniversitySapporoJapan
| | - Yuji Kumagai
- Division of Life Science, Graduate School of Life ScienceHokkaido UniversitySapporoJapan
| | - Seiichiro Ishihara
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life ScienceHokkaido UniversitySapporoJapan
| | - Atsushi Enomoto
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | | | - Motoaki Yasuda
- Department of Oral Pathobiological Science, Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Hisashi Haga
- Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life ScienceHokkaido UniversitySapporoJapan
| |
Collapse
|
15
|
From cyclins to CDKIs: Cell cycle regulation of skeletal muscle stem cell quiescence and activation. Exp Cell Res 2022; 420:113275. [PMID: 35931143 DOI: 10.1016/j.yexcr.2022.113275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/12/2022] [Accepted: 07/03/2022] [Indexed: 11/22/2022]
Abstract
After extensive proliferation during development, the adult skeletal muscle cells remain outside the cell cycle, either as post-mitotic myofibers or as quiescent muscle stem cells (MuSCs). Despite its terminally differentiated state, adult skeletal muscle has a remarkable regeneration potential, driven by MuSCs. Upon injury, MuSC quiescence is reversed to support tissue growth and repair and it is re-established after the completion of muscle regeneration. The distinct cell cycle states and transitions observed in the different myogenic populations are orchestrated by elements of the cell cycle machinery. This consists of i) complexes of cyclins and Cyclin-Dependent Kinases (CDKs) that ensure cell cycle progression and ii) their negative regulators, the Cyclin-Dependent Kinase Inhibitors (CDKIs). In this review we discuss the roles of these factors in developmental and adult myogenesis, with a focus on CDKIs that have emerging roles in stem cell functions.
Collapse
|
16
|
Identifying General Tumor and Specific Lung Cancer Biomarkers by Transcriptomic Analysis. BIOLOGY 2022; 11:biology11071082. [PMID: 36101460 PMCID: PMC9313083 DOI: 10.3390/biology11071082] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/25/2022] [Accepted: 07/03/2022] [Indexed: 11/17/2022]
Abstract
The bioinformatic pipeline previously developed in our research laboratory is used to identify potential general and specific deregulated tumor genes and transcription factors related to the establishment and progression of tumoral diseases, now comparing lung cancer with other two types of cancer. Twenty microarray datasets were selected and analyzed separately to identify hub differentiated expressed genes and compared to identify all the deregulated genes and transcription factors in common between the three types of cancer and those unique to lung cancer. The winning DEGs analysis allowed to identify an important number of TFs deregulated in the majority of microarray datasets, which can become key biomarkers of general tumors and specific to lung cancer. A coexpression network was constructed for every dataset with all deregulated genes associated with lung cancer, according to DAVID’s tool enrichment analysis, and transcription factors capable of regulating them, according to oPOSSUM´s tool. Several genes and transcription factors are coexpressed in the networks, suggesting that they could be related to the establishment or progression of the tumoral pathology in any tissue and specifically in the lung. The comparison of the coexpression networks of lung cancer and other types of cancer allowed the identification of common connectivity patterns with deregulated genes and transcription factors correlated to important tumoral processes and signaling pathways that have not been studied yet to experimentally validate their role in lung cancer. The Kaplan–Meier estimator determined the association of thirteen deregulated top winning transcription factors with the survival of lung cancer patients. The coregulatory analysis identified two top winning transcription factors networks related to the regulatory control of gene expression in lung and breast cancer. Our transcriptomic analysis suggests that cancer has an important coregulatory network of transcription factors related to the acquisition of the hallmarks of cancer. Moreover, lung cancer has a group of genes and transcription factors unique to pulmonary tissue that are coexpressed during tumorigenesis and must be studied experimentally to fully understand their role in the pathogenesis within its very complex transcriptomic scenario. Therefore, the downstream bioinformatic analysis developed was able to identify a coregulatory metafirm of cancer in general and specific to lung cancer taking into account the great heterogeneity of the tumoral process at cellular and population levels.
Collapse
|
17
|
Wang W, Tang W, Shan E, Zhang L, Chen S, Yu C, Gao Y. MiR-130a-5p contributed to the progression of endothelial cell injury by regulating FAS. Eur J Histochem 2022; 66. [PMID: 35638591 PMCID: PMC9201574 DOI: 10.4081/ejh.2022.3342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
MicroRNAs (miRNAs) play critical roles in the development of vascular diseases. However, the effects of miR-130a-5p and its functional targets on atherosclerosis (AS) are still largely unknown. In this regard, our aim is to explore the potentially important role of miR-130a-5p and its target gene during the progression of endothelial cell injury. We first found oxidized low-density lipoprotein (ox-LDL) induced FAS and cell apoptosis in HUVECs. Subsequently, miR-130a-5p expression was verified to be downregulated after ox-LDL treatment and negatively correlated with FAS, and FAS was identified as substantially upregulated in the ox-LDL-treated HUVEC cells. After that, the knockdown of FAS and overexpression of miR-130a-5p together were observed to aggregate ox-LDL-induced reduction of cell viability and apoptosis, cell cycle progression, cell proliferation, cell migration and invasion. In conclusion, we detected that miR-130a-5p contributed to the progression of endothelial cell injury by regulating of FAS, which may provide a new and promising therapeutic target for AS.
Collapse
|
18
|
Darnat P, Burg A, Sallé J, Lacoste J, Louvet-Vallée S, Gho M, Audibert A. Cortical Cyclin A controls spindle orientation during asymmetric cell divisions in Drosophila. Nat Commun 2022; 13:2723. [PMID: 35581185 PMCID: PMC9114397 DOI: 10.1038/s41467-022-30182-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 04/20/2022] [Indexed: 11/29/2022] Open
Abstract
The coordination between cell proliferation and cell polarity is crucial to orient the asymmetric cell divisions to generate cell diversity in epithelia. In many instances, the Frizzled/Dishevelled planar cell polarity pathway is involved in mitotic spindle orientation, but how this is spatially and temporally coordinated with cell cycle progression has remained elusive. Using Drosophila sensory organ precursor cells as a model system, we show that Cyclin A, the main Cyclin driving the transition to M-phase of the cell cycle, is recruited to the apical-posterior cortex in prophase by the Frizzled/Dishevelled complex. This cortically localized Cyclin A then regulates the orientation of the division by recruiting Mud, a homologue of NuMA, the well-known spindle-associated protein. The observed non-canonical subcellular localization of Cyclin A reveals this mitotic factor as a direct link between cell proliferation, cell polarity and spindle orientation. The Frizzled/Dishevelled planar cell polarity pathway is involved in mitotic spindle orientation, but how this is coordinated with the cell cycle is unclear. Here, the authors show with Drosophila sensory organ precursor cells that Cyclin A is recruited in prophase by Frizzled/Dishevelled, regulating division orientation.
Collapse
Affiliation(s)
- Pénélope Darnat
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Angélique Burg
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Jérémy Sallé
- Institut Jacques Monod, Université Paris Diderot/CNRS, Cellular Spatial Organization Team, F-75005, Paris, France
| | - Jérôme Lacoste
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Sophie Louvet-Vallée
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France
| | - Michel Gho
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France.
| | - Agnès Audibert
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement - Institut de Biologie Paris Seine (LBD-IBPS), Cell cycle and cell determination Team, F-75005, Paris, France.
| |
Collapse
|
19
|
Syahirah R, Hsu AY, Deng Q. A curious case of cyclin‐dependent kinases in neutrophils. J Leukoc Biol 2022; 111:1057-1068. [PMID: 35188696 PMCID: PMC9035055 DOI: 10.1002/jlb.2ru1021-573r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are terminally differentiated, short-lived white blood cells critical for innate immunity. Although cyclin-dependent kinases (CDKs) are typically related to cell cycle progression, increasing evidence has shown that they regulate essential functions of neutrophils. This review highlights the roles of CDKs and their partners, cyclins, in neutrophils, outside of cell cycle regulation. CDK1-10 and several cyclins are expressed in neutrophils, albeit at different levels. Observed phenotypes associated with specific inhibition or genetic loss of CDK2 indicate its role in modulating neutrophil migration. CDK4 and 6 regulate neutrophil extracellular traps (NETs) formation, while CDK5 regulates neutrophil degranulation. CDK7 and 9 are critical in neutrophil apoptosis, contributing to inflammation resolution. In addition to the CDKs that regulate mature neutrophil functions, cyclins are essential in hematopoiesis and granulopoiesis. The pivotal roles of CDKs in neutrophils present an untapped potential in targeting CDKs for treating neutrophil-dominant inflammatory diseases and understanding the regulation of the neutrophil life cycle.
Collapse
Affiliation(s)
- Ramizah Syahirah
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
| | - Alan Y. Hsu
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
- Department of Pathology Harvard Medical School Boston Massachusetts USA
- Department of Laboratory Medicine The Stem Cell Program, Boston Children's Hospital Boston Massachusetts USA
| | - Qing Deng
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
- Purdue Institute of Inflammation Immunology and Infectious Disease, Purdue University West Lafayette Indiana USA
- Purdue University Center for Cancer Research, Purdue University West Lafayette Indiana USA
| |
Collapse
|
20
|
Li S, Zhao J, Lv L, Dong D. Identification and Validation of TYMS as a Potential Biomarker for Risk of Metastasis Development in Hepatocellular Carcinoma. Front Oncol 2021; 11:762821. [PMID: 34858842 PMCID: PMC8630669 DOI: 10.3389/fonc.2021.762821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the major cause of hepatocellular carcinoma (HCC) mortality. Unfortunately, there are few reports on effective biomarkers for HCC metastasis. This study aimed to discover potential key genes of HCC, which could provide new insights for HCC metastasis. GEO (Gene Expression Omnibus) microarray and TCGA (The Cancer Genome Atlas) datasets were integrated to screen for candidate genes involved in HCC metastasis. Differentially expressed genes (DEGs) were screened, and then we performed enrichment analysis of Gene Ontology (GO), together with Kyoto Encyclopedia of Genes and Genomes (KEGG). A protein-protein interaction network was then built and analyzed utilizing STRING and Cytoscape, followed by the identification of 10 hub genes by cytoHubba. Four genes were associated with survival, their prognostic value was verified by prognostic signature analysis. Thymidylate synthase (TYMS) gene was identified as significant HCC metastasis-associated genes after mRNA expression validation and IHC analysis. TYMS silencing in HCC cells remarkedly inhibited growth and invasion. Finally, we found TYMS silencing dramatically decrease DNA synthesis and extracellular matrix (ECM) degradation, resulting in the inhibition of HCC metastasis, indicating TYMS had close associations with HCC development. These findings provided new insights into HCC metastasis and identified candidate gene prognosis signatures for HCC metastasis.
Collapse
Affiliation(s)
- Shuai Li
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingyuan Zhao
- Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Linlin Lv
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Atypical Teratoid Rhabdoid Tumours Are Susceptible to Panobinostat-Mediated Differentiation Therapy. Cancers (Basel) 2021; 13:cancers13205145. [PMID: 34680294 PMCID: PMC8534272 DOI: 10.3390/cancers13205145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Atypical teratoid rhabdoid tumour (ATRT) is an aggressive undifferentiated malignancy of the central nervous system in children. A defining feature of ATRT is the loss of the SMARCB1 gene that is essential for regulating gene expression required for normal developmental processes. We show that treatment of human ATRT cell models with the histone deacetylate inhibitor, panobinostat, inhibits tumour growth, reactivates the expression of developmental genes, and drives neuronal differentiation. These results demonstrate the therapeutic potential of panobinostat for the treatment of ATRT. Abstract Atypical teratoid rhabdoid tumour (ATRT) is a rare but highly aggressive undifferentiated solid tumour arising in the central nervous system and predominantly affecting infants and young children. ATRT is exclusively characterized by the inactivation of SMARCB1, a member of the SWI/SNF chromatin remodelling complex that is essential for the regulation of large sets of genes required for normal development and differentiation. Histone deacetylase inhibitors (HDACi) are a promising anticancer therapy and are able to mimic the normal acetylation functions of SMARCB1 in SMARCB1-deficient cells and drive multilineage differentiation in extracranial rhabdoid tumours. However, the potential efficacy of HDACi in ATRT is unknown. Here, we show that human ATRT cells are highly responsive to the HDACi panobinostat and that sustained treatment leads to growth arrest, increased cell senescence, decreased clonogenicity and induction of a neurogenesis gene-expression profile. Furthermore, in an orthotopic ATRT xenograft model, continuous panobinostat treatment inhibits tumour growth, increases survival and drives neuronal differentiation as shown by the expression of the neuronal marker, TUJ1. Collectively, this preclinical study supports the therapeutic potential of panobinostat-mediated differentiation therapy for ATRT.
Collapse
|
22
|
Lysophosphatidic Acid Mediates Imiquimod-Induced Psoriasis-like Symptoms by Promoting Keratinocyte Proliferation through LPAR1/ROCK2/PI3K/AKT Signaling Pathway. Int J Mol Sci 2021; 22:ijms221910777. [PMID: 34639115 PMCID: PMC8509620 DOI: 10.3390/ijms221910777] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease. Recently, lysophosphatidic acid (LPA)/LPAR5 signaling has been reported to be involved in both NLRP3 inflammasome activation in macrophages and keratinocyte activation to produce inflammatory cytokines, contributing to psoriasis pathogenesis. However, the effect and molecular mechanisms of LPA/LPAR signaling in keratinocyte proliferation in psoriasis remain unclear. In this study, we investigated the effects of LPAR1/3 inhibition on imiquimod (IMQ)-induced psoriasis-like mice. Treatment with the LPAR1/3 antagonist, ki16425, alleviated skin symptoms in IMQ-induced psoriasis-like mouse models and decreased keratinocyte proliferation in the lesion. It also decreased LPA-induced cell proliferation and cell cycle progression via increased cyclin A2, cyclin D1, cyclin-dependent kinase (CDK)2, and CDK4 expression and decreased p27Kip1 expression in HaCaT cells. LPAR1 knockdown in HaCaT cells reduced LPA-induced proliferation, suppressed cyclin A2 and CDK2 expression, and restored p27Kip1 expression. LPA increased Rho-associated protein kinase 2 (ROCK2) expression and PI3K/AKT activation; moreover, the pharmacological inhibition of ROCK2 and PI3K/AKT signaling suppressed LPA-induced cell cycle progression. In conclusion, we demonstrated that LPAR1/3 antagonist alleviates IMQ-induced psoriasis-like symptoms in mice, and in particular, LPAR1 signaling is involved in cell cycle progression via ROCK2/PI3K/AKT pathways in keratinocytes.
Collapse
|
23
|
Suski JM, Braun M, Strmiska V, Sicinski P. Targeting cell-cycle machinery in cancer. Cancer Cell 2021; 39:759-778. [PMID: 33891890 PMCID: PMC8206013 DOI: 10.1016/j.ccell.2021.03.010] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/09/2021] [Accepted: 03/26/2021] [Indexed: 12/19/2022]
Abstract
Abnormal activity of the core cell-cycle machinery is seen in essentially all tumor types and represents a driving force of tumorigenesis. Recent studies revealed that cell-cycle proteins regulate a wide range of cellular functions, in addition to promoting cell division. With the clinical success of CDK4/6 inhibitors, it is becoming increasingly clear that targeting individual cell-cycle components may represent an effective anti-cancer strategy. Here, we discuss the potential of inhibiting different cell-cycle proteins for cancer therapy.
Collapse
Affiliation(s)
- Jan M Suski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Marcin Braun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Chair of Oncology, Medical University of Lodz, 92-213 Lodz, Poland
| | - Vladislav Strmiska
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Lu XQ, Zhang JQ, Zhang SX, Qiao J, Qiu MT, Liu XR, Chen XX, Gao C, Zhang HH. Identification of novel hub genes associated with gastric cancer using integrated bioinformatics analysis. BMC Cancer 2021; 21:697. [PMID: 34126961 PMCID: PMC8201699 DOI: 10.1186/s12885-021-08358-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 05/13/2021] [Indexed: 02/07/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common solid malignant tumors worldwide with a high-recurrence-rate. Identifying the molecular signatures and specific biomarkers of GC might provide novel clues for GC prognosis and targeted therapy. Methods Gene expression profiles were obtained from the ArrayExpress and Gene Expression Omnibus database. Differentially expressed genes (DEGs) were picked out by R software. The hub genes were screened by cytohubba plugin. Their prognostic values were assessed by Kaplan–Meier survival analyses and the gene expression profiling interactive analysis (GEPIA). Finally, qRT-PCR in GC tissue samples was established to validate these DEGs. Results Total of 295 DEGs were identified between GC and their corresponding normal adjacent tissue samples in E-MTAB-1440, GSE79973, GSE19826, GSE13911, GSE27342, GSE33335 and GSE56807 datasets, including 117 up-regulated and 178 down-regulated genes. Among them, 7 vital upregulated genes (HMMR, SPP1, FN1, CCNB1, CXCL8, MAD2L1 and CCNA2) were selected. Most of them had a significantly worse prognosis except SPP1. Using qRT-PCR, we validated that their transcriptions in our GC tumor tissue were upregulated except SPP1 and FN1, which correlated with tumor relapse and predicts poorer prognosis in GC patients. Conclusions We have identified 5 upregulated DEGs (HMMR, CCNB1, CXCL8, MAD2L1, and CCNA2) in GC patients with poor prognosis using integrated bioinformatical methods, which could be potential biomarkers and therapeutic targets for GC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08358-7.
Collapse
Affiliation(s)
- Xiao-Qing Lu
- Department of Breast Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Jia-Qian Zhang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jun Qiao
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meng-Ting Qiu
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiang-Rong Liu
- Department of Breast Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Xiao-Xia Chen
- Department of Breast Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Huan-Hu Zhang
- Department of Gastroenterology, Shanxi Cancer Hospital, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
25
|
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: 2.3] [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
|
26
|
Simulated Microgravity Inhibits the Proliferation of Chang Liver Cells by Attenuation of the Major Cell Cycle Regulators and Cytoskeletal Proteins. Int J Mol Sci 2021; 22:ijms22094550. [PMID: 33925309 PMCID: PMC8123698 DOI: 10.3390/ijms22094550] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 11/29/2022] Open
Abstract
Simulated microgravity (SMG) induced the changes in cell proliferation and cytoskeleton organization, which plays an important factor in various cellular processes. The inhibition in cell cycle progression has been considered to be one of the main causes of proliferation inhibition in cells under SMG, but their mechanisms are still not fully understood. This study aimed to evaluate the effects of SMG on the proliferative ability and cytoskeleton changes of Chang Liver Cells (CCL-13). CCL-13 cells were induced SMG by 3D clinostat for 72 h, while the control group were treated in normal gravity at the same time. The results showed that SMG reduced CCL-13 cell proliferation by an increase in the number of CCL-13 cells in G0/G1 phase. This cell cycle phase arrest of CCL-13 cells was due to a downregulation of cell cycle-related proteins, such as cyclin A1 and A2, cyclin D1, and cyclin-dependent kinase 6 (Cdk6). SMG-exposed CCL-13 cells also exhibited a downregulation of α-tubulin 3 and β-actin which induced the cytoskeleton reorganization. These results suggested that the inhibited proliferation of SMG-exposed CCL-13 cells could be associate with the attenuation of major cell cycle regulators and main cytoskeletal proteins.
Collapse
|
27
|
Jiang P, Zhang M, Gui L, Zhang K. Expression patterns and prognostic values of the cyclin-dependent kinase 1 and cyclin A2 gene cluster in pancreatic adenocarcinoma. J Int Med Res 2021; 48:300060520930113. [PMID: 33290118 PMCID: PMC7727076 DOI: 10.1177/0300060520930113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Objective Pancreatic adenocarcinoma (PAAD) is one of the most lethal malignant tumors worldwide. Various studies based on cell lines, preclinical mouse models, and human tissue samples have shown that cell cycle-associated proteins are involved in the tumorigenesis and progression of PAAD. Methods Herein, we analyzed the relationships between CDK1 and CCNA2 gene expression and prognosis in patients with pancreatic cancer, using information from the Oncomine, cBioportal, Kaplan–Meier Plotter, and GEPIA databases. Results Expression levels of CDK1 and CCNA2 were significantly higher in PAAD compared with control tissues, and were associated with more advanced tumor stage. Survival analyses using the Kaplan–Meier Plotter database further confirmed that increased expression levels of CDK1 and CCNA2 were associated with a poor prognosis in patients with pancreatic cancer. Conclusions The results of this study suggest that CDK1 and CCNA2 may be potential therapeutic targets and prognostic biomarkers in patients with PAAD.
Collapse
Affiliation(s)
- Peng Jiang
- Department of Gastroenterology, The Central Hospital of Weihai, Weihai, Shandong, China
| | - Ming Zhang
- Hepatobiliary Surgery Department, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Liangliang Gui
- Department of Gastroenterology, The Central Hospital of Weihai, Weihai, Shandong, China
| | - Kai Zhang
- Hepatobiliary Surgery Department, Shandong Provincial Third Hospital, Jinan, Shandong, China
| |
Collapse
|
28
|
Wang G, Li Q, Li C, Duan G, Sang H, Dong H, Yang Y, Ma C, Tao T. Knockdown of PNO1 inhibits esophageal cancer progression. Oncol Rep 2021; 45:85. [PMID: 33864661 PMCID: PMC8025143 DOI: 10.3892/or.2021.8036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022] Open
Abstract
The present study aimed to investigate the role of partner of NOB1 homolog (PNO1) in esophageal cancer (EC). The expression levels of PNO1 in EC were primarily analyzed using data obtained from databases. PNO1 expression was also knocked down in EC cells (Eca-109 and TE1) to determine the biological effects of PNO1 on tumorigenesis in vitro and in vivo. In addition, possible downstream targets of PNO1 in EC were identified. The expression levels of PNO1 were upregulated in the tumor tissues compared with that noted in normal tissues. Moreover, the knockdown (KD) of PNO1 suppressed cell proliferation, migration and invasion, and promoted cell apoptosis (P<0.05). Furthermore, the protein expression levels of AKT1, Twist, Myc, mTOR, matrix metalloproteinase 2 (MMP2), nuclear factor (NF)-κB p65 and β-catenin 1 (CTNNB1) were downregulated following the KD of PNO1 in Eca-109 cells (P<0.05). In addition, the overexpression of CTNNB1 reversed the effects of PNO1 KD in Eca-109 cells (P<0.05). In conclusion, the findings of the present study suggest that PNO1 promotes EC progression by regulating AKT1, Twist, Myc, mTOR, MMP2, NF-κB p65 and CTNNB1 expression.
Collapse
Affiliation(s)
- Guowen Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Qicai Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chuankui Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Guixin Duan
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Haiwei Sang
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Haijun Dong
- Department of Thoracic Surgery, Huzhou Central Hospital, Huzhou, Zhejiang 310000, P.R. China
| | - Yifan Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chang Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Tao Tao
- Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
29
|
Silva Cascales H, Burdova K, Middleton A, Kuzin V, Müllers E, Stoy H, Baranello L, Macurek L, Lindqvist A. Cyclin A2 localises in the cytoplasm at the S/G2 transition to activate PLK1. Life Sci Alliance 2021; 4:e202000980. [PMID: 33402344 PMCID: PMC7812317 DOI: 10.26508/lsa.202000980] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 01/23/2023] Open
Abstract
Cyclin A2 is a key regulator of the cell cycle, implicated both in DNA replication and mitotic entry. Cyclin A2 participates in feedback loops that activate mitotic kinases in G2 phase, but why active Cyclin A2-CDK2 during the S phase does not trigger mitotic kinase activation remains unclear. Here, we describe a change in localisation of Cyclin A2 from being only nuclear to both nuclear and cytoplasmic at the S/G2 border. We find that Cyclin A2-CDK2 can activate the mitotic kinase PLK1 through phosphorylation of Bora, and that only cytoplasmic Cyclin A2 interacts with Bora and PLK1. Expression of predominately cytoplasmic Cyclin A2 or phospho-mimicking PLK1 T210D can partially rescue a G2 arrest caused by Cyclin A2 depletion. Cytoplasmic presence of Cyclin A2 is restricted by p21, in particular after DNA damage. Cyclin A2 chromatin association during DNA replication and additional mechanisms contribute to Cyclin A2 localisation change in the G2 phase. We find no evidence that such mechanisms involve G2 feedback loops and suggest that cytoplasmic appearance of Cyclin A2 at the S/G2 transition functions as a trigger for mitotic kinase activation.
Collapse
Affiliation(s)
| | - Kamila Burdova
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Anna Middleton
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Vladislav Kuzin
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Erik Müllers
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Henriette Stoy
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Laura Baranello
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Libor Macurek
- Laboratory of Cancer Cell Biology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Arne Lindqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
30
|
Kim SS, Alves MJ, Gygli P, Otero J, Lindert S. Identification of Novel Cyclin A2 Binding Site and Nanomolar Inhibitors of Cyclin A2-CDK2 Complex. Curr Comput Aided Drug Des 2021; 17:57-68. [PMID: 31889491 DOI: 10.2174/1573409916666191231113055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/25/2019] [Accepted: 12/09/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Given the diverse roles of cyclin A2 both in cell cycle regulation and in DNA damage response, identifying small molecule regulators of cyclin A2 activity carries significant potential to regulate diverse cellular processes in both ageing/neurodegeneration and in cancer. OBJECTIVE Based on cyclin A2's recently discovered role in DNA repair, we hypothesized that small molecule inhibitors that were predicted to bind to both cyclin A2 and CDK2 will be useful as a radiosensitizer of cancer cells. In this study, we used structure-based drug discovery to find inhibitors that target both cyclin A2 and CDK2. METHODS Molecular dynamics simulations were used to generate diverse binding pocket conformations for application of the relaxed complex scheme. We then used structure-based virtual screening to find potential dual cyclin A2 and CDK2 inhibitors. Based on a consensus score of docked poses from Glide and AutoDock Vina, we identified about 40 promising hit compounds, where all PAINS scaffolds were removed from consideration. A biochemical luminescence assay of cyclin A2-CDK2 function was used for experimental verification. RESULTS Four lead inhibitors of cyclin A2-CDK2 complex have been identified using a relaxed complex scheme virtual screen have been verified in a biochemical luminescence assay of cyclin A2- CDK2 function. Two of the four lead inhibitors had inhibitory concentrations in the nanomolar range. CONCLUSION The four cyclin A2-CDK2 complex inhibitors are the first reported inhibitors that were specifically designed not to target the cyclin A2-CDK2 protein-protein interface. Overall, our results highlight the potential of combined advanced computational tools and biochemical verification to discover novel binding scaffolds.
Collapse
Affiliation(s)
- Stephanie S Kim
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, 43210, United States
| | - Michele J Alves
- Departments of Neuroscience, Pathology and Neuropathology, Ohio State University, Columbus, OH, 43210, United States
| | - Patrick Gygli
- Departments of Neuroscience, Pathology and Neuropathology, Ohio State University, Columbus, OH, 43210, United States
| | - Jose Otero
- Departments of Neuroscience, Pathology and Neuropathology, Ohio State University, Columbus, OH, 43210, United States
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, 43210, United States
| |
Collapse
|
31
|
Guo Y, Gabola M, Lattanzio R, Paul C, Pinet V, Tang R, Turali H, Bremond J, Longobardi C, Maurizy C, Da Costa Q, Finetti P, Boissière-Michot F, Rivière B, Lemmers C, Garnier S, Bertucci F, Zlobec I, Chebli K, Tazi J, Azar R, Blanchard JM, Sicinski P, Mamessier E, Lemmers B, Hahne M. Cyclin A2 maintains colon homeostasis and is a prognostic factor in colorectal cancer. J Clin Invest 2021; 131:131517. [PMID: 33332285 DOI: 10.1172/jci131517] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
To clarify the function of cyclin A2 in colon homeostasis and colorectal cancer (CRC), we generated mice deficient for cyclin A2 in colonic epithelial cells (CECs). Colons of these mice displayed architectural changes in the mucosa and signs of inflammation, as well as increased proliferation of CECs associated with the appearance of low- and high-grade dysplasias. The main initial events triggering those alterations in cyclin A2-deficient CECs appeared to be abnormal mitoses and DNA damage. Cyclin A2 deletion in CECs promoted the development of dysplasia and adenocarcinomas in a murine colitis-associated cancer model. We next explored the status of cyclin A2 expression in clinical CRC samples at the mRNA and protein levels and found higher expression in tumors of patients with stage 1 or 2 CRC compared with those of patients with stage 3 or 4 CRC. A meta-analysis of 11 transcriptome data sets comprising 2239 primary CRC tumors revealed different expression levels of CCNA2 (the mRNA coding for cyclin A2) among the CRC tumor subtypes, with the highest expression detected in consensus molecular subtype 1 (CMS1) and the lowest in CMS4 tumors. Moreover, we found high expression of CCNA2 to be a new, independent prognosis factor for CRC tumors.
Collapse
Affiliation(s)
- Yuchen Guo
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Monica Gabola
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Rossano Lattanzio
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University, Chieti, Italy.,Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Conception Paul
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Valérie Pinet
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Ruizhi Tang
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Hulya Turali
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Julie Bremond
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Ciro Longobardi
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Chloé Maurizy
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Quentin Da Costa
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), INSERM, U1068, CNRS, UMR7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), INSERM, U1068, CNRS, UMR7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France
| | - Florence Boissière-Michot
- Translationnal Research Unit, Montpellier Cancer Institute, Montpellier, France - Université de Montpellier, Montpellier, France
| | - Benjamin Rivière
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Céline Lemmers
- PVM, Biocampus, Université de Montpellier, CNRS, Montpellier, France
| | - Séverine Garnier
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), INSERM, U1068, CNRS, UMR7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France
| | - François Bertucci
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), INSERM, U1068, CNRS, UMR7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France.,Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Karim Chebli
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Jamal Tazi
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Rania Azar
- Faculty of Pharmacy, Lebanese University, Hadath, Lebanon
| | - Jean-Marie Blanchard
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | | | - Emilie Mamessier
- Predictive Oncology Laboratory, Cancer Research Center of Marseille (CRCM), INSERM, U1068, CNRS, UMR7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France
| | - Bénédicte Lemmers
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Michael Hahne
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| |
Collapse
|
32
|
Wei LM, Li XY, Wang ZM, Wang YK, Yao G, Fan JH, Wang XS. Identification of hub genes in triple-negative breast cancer by integrated bioinformatics analysis. Gland Surg 2021; 10:799-806. [PMID: 33708561 DOI: 10.21037/gs-21-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Triple negative breast cancer (TNBC) is usually aggressive and accompanied by a poor prognosis. The molecular biological mechanism of TNBC pathogenesis is still unclear, and requires more detailed research. The aim of this study was to screen and verify potential biomarkers of TNBC, and provide new clues for the treatment and diagnosis of TNBC. Methods In this work, GSE76250 was downloaded from the Gene Expression Omnibus (GEO) database and included 165 TNBC samples and 33 paired normal breast tissues. The R software and its related software package were used for data processing and analysis. Compared with normal tissues, genes with a false discovery rate (FDR) <0.01 and log fold change (logFC) ≥1 or ≤-1 were identified as differentially expressed genes (DEGs) by limma package. Survival prognoses were analyzed by Kaplan-Meier plotter database. Results In total, 160 up-regulated and 180 down-regulated genes were identified. The biological mechanism of enrichment analysis presented that DEGs were significantly enriched in chromosome segregation, extracellular matrix, and extracellular matrix structural constituent, among others. A total of 8 hub genes (CCNB1, CDK1, TOP2A, MKI67, TTK, CCNA2, BUB1, and PLK1) were identified by the protein-protein interaction network (PPIN) and Cytoscape software. Survival prognosis of these hub genes showed that they were negatively correlated with overall survival. Conclusions The 8 hub genes and pathways that were identified might be involved in tumorigenesis and become new candidate biomarkers for TNBC treatment.
Collapse
Affiliation(s)
- Li-Min Wei
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xin-Yang Li
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Zi-Ming Wang
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yu-Kun Wang
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ge Yao
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jia-Hao Fan
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xin-Shuai Wang
- Department of Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
33
|
Gao L, Xia S, Zhang K, Lin C, He X, Zhang Y. Gene expression profile of THZ1-treated nasopharyngeal carcinoma cell lines indicates its involvement in the inhibition of the cell cycle. Transl Cancer Res 2021; 10:445-460. [PMID: 35116274 PMCID: PMC8799269 DOI: 10.21037/tcr-19-2888] [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: 12/20/2019] [Accepted: 09/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The aim of this study was to identify downstream target genes and pathways regulated by THZ1 in nasopharyngeal carcinoma (NPC). METHODS The gene expression profile of GSE95750 in two NPC cell lines, untreated group and treated with THZ1 group, was analyzed. Differentially expressed genes (DEGs) were compared using the R-software. Then Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathways (KEGG) was analyzed using Database for Annotation, Visualization, and Integrated Discovery (DAVID). Cytoscape was used for protein-protein interaction (PPI) analysis. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to verified the gene expression. RESULTS We identified 25 genes with increased expression and 567 genes with decreased expression in THZ1-treated NPC cells. The top 10 significantly DEGs between untreated group and THZ1 treated group were identified by qRT-PCR and the results were in agreement with RNA-seq. The total 592 DEGs were found enriched in 1,148 GO terms and 38 KEGG pathways. The most important enriched pathways identified were cell cycle related, and several related node genes were identified, such as CDC6, CDC34, CDK7, CDK9, CCNA2, CCNB1, CDT1, KIF11, LIN9, PLK1, and POLR family, which consistent with RNA-seq. CONCLUSIONS Our results emphasize the differential genes and pathways occurring in THZ1-treated NPC cells, which increases our understanding of the anti-tumor mechanisms of THZ1.
Collapse
Affiliation(s)
- Lijuan Gao
- Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Radiation Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Shuang Xia
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Kunyi Zhang
- Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Radiation Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Chengguang Lin
- Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Radiation Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Xuyu He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ying Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
34
|
Krajewski A, Gagat M, Mikołajczyk K, Izdebska M, Żuryń A, Grzanka A. Cyclin F Downregulation Affects Epithelial-Mesenchymal Transition Increasing Proliferation and Migration of the A-375 Melanoma Cell Line. Cancer Manag Res 2020; 12:13085-13097. [PMID: 33376401 PMCID: PMC7765751 DOI: 10.2147/cmar.s279169] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/19/2020] [Indexed: 01/22/2023] Open
Abstract
Background Cyclins are well-known cell cycle regulators. The activation of cyclin-dependent kinases by cyclins allows orchestration of the complicated cell cycle machinery and drives the cell from the G1 phase to the end of the mitotic phase. In recent years, it has become evident that cyclins are involved in processes beyond the cell cycle. Cyclin F does not activate CDKs but forms part of the Skp1-Cul1-F-box (SCF) complex where it is responsible for protein target recognition and subsequent degradation in a proteasome-dependent manner. Results Here, we report that the downregulation of cyclin F in the A-375 melanoma cell line increases cell viability and colony formation in a cell cycle independent manner. Lower levels of cyclin F do not appear to affect the cell cycle, based on flow cytometry measuring BrdU incorporation and propidium iodide staining. By means of immunofluorescence staining and Western blot analysis, we observed changes in cell morphology-related markers which suggested ongoing epithelial-mesenchymal transition (EMT) in response to cyclin F downregulation. Increases in vimentin and N-cadherin protein levels, decreases in levels of epithelial markers such as ZO-1, along with changes in morphology to a spindle-like shape with the appearance of actin stress fibers, are all hallmarks of EMT. These changes are associated with increased invasive and migratory potential, based on 2D migration assays. Moreover, we observe an increase in RhoABC, talin and paxillin levels, the proteins involved in controlling cell signaling and motility. Lastly, upon knocking down cyclin F expression, we observed a decrease in thrombospondin-1 expression, suggesting a role of cyclin F in angiogenesis. Conclusion Cyclin F depletion induces proliferation and EMT processes in the A-375 melanoma model.
Collapse
Affiliation(s)
- Adrian Krajewski
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | - Maciej Gagat
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | - Klaudia Mikołajczyk
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | - Magdalena Izdebska
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | - Agnieszka Żuryń
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| |
Collapse
|
35
|
Establishment of a Gene Signature to Predict Prognosis for Patients with Lung Adenocarcinoma. Int J Mol Sci 2020; 21:ijms21228479. [PMID: 33187219 PMCID: PMC7697394 DOI: 10.3390/ijms21228479] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence indicates that the reliable gene signature may serve as an independent prognosis factor for lung adenocarcinoma (LUAD) diagnosis. Here, we sought to identify a risk score signature for survival prediction of LUAD patients. In the Gene Expression Omnibus (GEO) database, GSE18842, GSE75037, GSE101929, and GSE19188 mRNA expression profiles were downloaded to screen differentially expressed genes (DEGs), which were used to establish a protein-protein interaction network and perform clustering module analysis. Univariate and multivariate proportional hazards regression analyses were applied to develop and validate the gene signature based on the TCGA dataset. The signature genes were then verified on GEPIA, Oncomine, and HPA platforms. Expression levels of corresponding genes were also measured by qRT-PCR and Western blotting in HBE, A549, and PC-9 cell lines. The prognostic signature based on eight genes (TTK, HMMR, ASPM, CDCA8, KIF2C, CCNA2, CCNB2, and MKI67) was established, which was independent of other clinical factors. The risk model offered better discrimination between risk groups, and patients with high-risk scores tended to have poor survival rate at 1-, 3- and 5-year follow-up. The model also presented better survival prediction in cancer-specific cohorts of age, gender, clinical stage III/IV, primary tumor 1/2, and lymph node metastasis 1/2. The signature genes, moreover, were highly expressed in A549 and PC-9 cells. In conclusion, the risk score signature could be used for prognostic estimation and as an independent risk factor for survival prediction in patients with LUAD.
Collapse
|
36
|
Naorem LD, Pathak E, Muthaiyan M, Venkatesan A. Network-based meta-analysis for the identification of potential target for human anaplastic thyroid carcinoma. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
37
|
Ma F, Wang X, Chung SSW, Sicinski P, Shang E, Wolgemuth DJ. Cyclin A2 is essential for mouse gonocyte maturation. Cell Cycle 2020; 19:1654-1664. [PMID: 32420805 DOI: 10.1080/15384101.2020.1762314] [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] [Indexed: 10/24/2022] Open
Abstract
In mammals, male gonocytes are derived from primordial germ cells during embryogenesis, enter a period of mitotic proliferation, and then become quiescent until birth. After birth, the gonocytes proliferate and migrate from the center of testicular cord toward the basement membrane to form the pool of spermatogonial stem cells (SSCs) and establish the SSC niche architecture. However, the molecular mechanisms underlying gonocyte proliferation, migration and differentiation are largely unknown. Cyclin A2 is a key component of the cell cycle and required for cell proliferation. Here, we show that cyclin A2 is required in mouse male gonocyte development and the establishment of spermatogenesis in the neonatal testis. Loss of cyclin A2 function in embryonic gonocytes by targeted gene disruption affected the regulation of the male gonocytes to SSC transition, resulting in the disruption of SSC pool formation, imbalance between SSC self-renewal and differentiation, and severely abnormal spermatogenesis in the adult testis.
Collapse
Affiliation(s)
- Fanhua Ma
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction, College of Animal Science, Huazhong Agricultural University , Wuhan, Hubei, China.,Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA
| | - Xiangyuan Wang
- Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA
| | - Sanny S W Chung
- Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute , Boston, MA, USA
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York , New York, NY, USA
| | - Debra J Wolgemuth
- Department of Genetics & Development, Columbia University Medical Center , New York, NY, USA.,Institute of Human Nutrition, Columbia University Medical Center , New York, NY, USA
| |
Collapse
|
38
|
Ma X, Ren H, Peng R, Li Y, Ming L. Identification of key genes associated with progression and prognosis for lung squamous cell carcinoma. PeerJ 2020; 8:e9086. [PMID: 32411535 PMCID: PMC7210810 DOI: 10.7717/peerj.9086] [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: 09/27/2019] [Accepted: 04/08/2020] [Indexed: 12/24/2022] Open
Abstract
Background Lung squamous cell carcinoma (LUSC) is a major subtype of lung cancer with limited therapeutic options and poor clinical prognosis. Methods Three datasets (GSE19188, GSE33532 and GSE33479) were obtained from the gene expression omnibus (GEO) database. Differentially expressed genes (DEGs) between LUSC and normal tissues were identified by GEO2R, and functional analysis was employed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool. Protein-protein interaction (PPI) and hub genes were identified via the Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape software. Hub genes were further validated in The Cancer Genome Atlas (TCGA) database. Subsequently, survival analysis was performed using the Kapla-Meier curve and Cox progression analysis. Based on univariate and multivariate Cox progression analysis, a gene signature was established to predict overall survival. Receiver operating characteristic curve was used to evaluate the prognostic value of the model. Results A total of 116 up-regulated genes and 84 down-regulated genes were identified. These DEGs were mainly enriched in the two pathways: cell cycle and p53 signaling way. According to the degree of protein nodes in the PPI network, 10 hub genes were identified. The mRNA expression levels of the 10 hub genes in LUSC were also significantly up-regulated in the TCGA database. Furthermore, a novel seven-gene signature (FLRT3, PPP2R2C, MMP3, MMP12, CAPN8, FILIP1 and SPP1) from the DEGs was constructed and acted as a significant and independent prognostic signature for LUSC. Conclusions The 10 hub genes might be tightly correlated with LUSC progression. The seven-gene signature might be an independent biomarker with a significant predictive value in LUSC overall survival.
Collapse
Affiliation(s)
- Xiaohan Ma
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Huijun Ren
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Ruoyu Peng
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Yi Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| | - Liang Ming
- Department of Clinical Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Clinical Laboratory of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
39
|
Lu Y, Yang G, Xiao Y, Zhang T, Su F, Chang R, Ling X, Bai Y. Upregulated cyclins may be novel genes for triple-negative breast cancer based on bioinformatic analysis. Breast Cancer 2020; 27:903-911. [PMID: 32338339 DOI: 10.1007/s12282-020-01086-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 04/02/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the leading causes of death among females around the world. However, the molecular mechanism of the disease among TNBC patients remains to be further studied. METHODS In our study, four microarray data and two high throughput sequencing data were acquired from the GEO database, and the differentially expressed genes (DEGs) between TNBC and normal tissues had been analyzed. Analysis of functional enrichment and pathway enrichment of DEGs was conducted by the Funrich software, and protein-protein interaction (PPI) network gained from the STRING, and hub genes were confirmed by the Cytoscape. Kaplan-Meier plotter (KM plotter) online dataset had been used to analyze DEGs of overall survival (OS), and progression-free survival (PFS). RESULTS In total, 1638 DEGs were gained in our study covering 984 upregulated and 654 downregulated genes. Moreover, a PPI network was constructed, and cyclin-dependent kinase 1 (CDK1), cyclin B1 (CCNB1), and cyclin A2 (CCNA2) were found as top genes with higher node degrees. CDK1, CCNA2, and CCNB1were obviously enriched in the cell cycle. The top upregulated genes including CDK1, CCNB1, CCNA2, and PLK1 were overexpressed in TNBC, and correlated with worse OS in breast cancer. High expression of CCNB1 was correlated with worse PFS in TNBC (HR = 1.42, 95% CI: 1.04-1.94, P = 0.028). Besides, there was a correlation between CCNB1 and CDK1 in TNBC, as well as between CCNA2 and CDK1 (r = 0.804, P < 0.001; r = 0.577, P < 0.001, respectively). CONCLUSION Our results suggest that cyclin CDK1, CCNB1, and CCNA2 are overexpressed in TNBC and they could act as novel biomarkers for the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Yongbin Lu
- Scientific Development and Planning Department, The First Hospital of Lanzhou University, Lanzhou, China
- College of Earth and Environmental Sciences, Lanzhou University, Lanzhou, China
| | - Gang Yang
- Neurosurgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yi Xiao
- Breast Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Tao Zhang
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Fei Su
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Ruixia Chang
- School of Public Health, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoling Ling
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, 730000, People's Republic of China.
| | - Yana Bai
- College of Earth and Environmental Sciences, Lanzhou University, Lanzhou, China.
- Epidemiology and Health Statistics, School of Public Health, Lanzhou University, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
40
|
Farahani M, Rezaei‐Tavirani M, Zali H, Hatamie S, Ghasemi N. Systems toxicology assessment revealed the impact of graphene‐based materials on cell cycle regulators. J Biomed Mater Res A 2020; 108:1520-1533. [DOI: 10.1002/jbm.a.36923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Masoumeh Farahani
- Proteomics Research CenterShahid Beheshti University of Medical Sciences Tehran Iran
| | | | - Hakimeh Zali
- Proteomics Research CenterShahid Beheshti University of Medical Sciences Tehran Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineShahid Beheshti University of Medical Sciences Tehran Iran
| | - Shadie Hatamie
- Institute of NanoEngineering and MicroSystemsNational Tsing Hua University Hsinchu Taiwan
- Department of Power Mechanical EngineeringNational Tsing Hua University Hsinchu Taiwan
| | - Nazanin Ghasemi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineShahid Beheshti University of Medical Sciences Tehran Iran
- Department of Immunology, School of MedicineShahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
41
|
Ali A, Al-Tobasei R, Lourenco D, Leeds T, Kenney B, Salem M. Genome-wide identification of loci associated with growth in rainbow trout. BMC Genomics 2020; 21:209. [PMID: 32138655 PMCID: PMC7059289 DOI: 10.1186/s12864-020-6617-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background Growth is a major economic production trait in aquaculture. Improvements in growth performance will reduce time and cost for fish to reach market size. However, genes underlying growth have not been fully explored in rainbow trout. Results A previously developed 50 K gene-transcribed SNP chip, containing ~ 21 K SNPs showing allelic imbalances potentially associated with important aquaculture production traits including body weight, muscle yield, was used for genotyping a total of 789 fish with available phenotypic data for bodyweight gain. Genotyped fish were obtained from two consecutive generations produced in the NCCCWA growth-selection breeding program. Weighted single-step GBLUP (WssGBLUP) was used to perform a genome-wide association (GWA) analysis to identify quantitative trait loci (QTL) associated with bodyweight gain. Using genomic sliding windows of 50 adjacent SNPs, 247 SNPs associated with bodyweight gain were identified. SNP-harboring genes were involved in cell growth, cell proliferation, cell cycle, lipid metabolism, proteolytic activities, chromatin modification, and developmental processes. Chromosome 14 harbored the highest number of SNPs (n = 50). An SNP window explaining the highest additive genetic variance for bodyweight gain (~ 6.4%) included a nonsynonymous SNP in a gene encoding inositol polyphosphate 5-phosphatase OCRL-1. Additionally, based on a single-marker GWA analysis, 33 SNPs were identified in association with bodyweight gain. The highest SNP explaining variation in bodyweight gain was identified in a gene coding for thrombospondin-1 (THBS1) (R2 = 0.09). Conclusion The majority of SNP-harboring genes, including OCRL-1 and THBS1, were involved in developmental processes. Our results suggest that development-related genes are important determinants for growth and could be prioritized and used for genomic selection in breeding programs.
Collapse
Affiliation(s)
- Ali Ali
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Rafet Al-Tobasei
- Computational Science Program, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Daniela Lourenco
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Tim Leeds
- United States Department of Agriculture Kearneysville, National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, Kearneysville, WV, USA
| | - Brett Kenney
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Mohamed Salem
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
42
|
Jiang Y, Hong D, Lou Z, Tu X, Jin L. Lupeol inhibits migration and invasion of colorectal cancer cells by suppressing RhoA-ROCK1 signaling pathway. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:2185-2196. [PMID: 32025757 DOI: 10.1007/s00210-020-01815-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 01/08/2020] [Indexed: 12/19/2022]
Abstract
Metastasis is the main cause of death in colorectal cancer (CRC) patients. However, current treatment options for CRC metastasis are very limited. Lupeol, a triterpene that is widely found in vegetables and fruits, has been reported to possess the cancer-preventive and anti-inflammatory functions. However, the roles of Lupeol in the migration and invasion of colorectal cancer remain unclear. Here, we evaluated the effect of Lupeol treatment on colorectal cancer cell lines, HCT116 and SW620, and delineated its underlying mechanisms. Our results showed that Lupeol induced a dose-dependent inhibition of HCT116 and SW620 cells viability, measured by CCK8 assay. Wound healing and Transwell migration and invasion assays revealed that Lupeol significantly suppressed the migration and invasion of CRC cells. Using laser confocal microscope, we observed that the pseudopods and protrusions of HCT116 and SW620 cells decreased and disrupted after treatment with Lupeol. In addition, the quantitative real-time PCR and Western blotting results showed that Lupeol downregulated the expression of RhoA and RhoC, and their downstream effectors ROCK1, Cofilin, p-MLC, and the associated regulatory protein Cyclin A2. Interestingly, the migration and invasion capacity of CRC cells was reduced after RhoA knockdown. And there were no additional changes in CRC cells with RhoA knockdown to treat with Lupeol. These findings demonstrate that Lupeol can suppress the migration and invasion of colorectal cancer cells by remodeling the actin cytoskeleton via RhoA-ROCK1 pathway inhibition, which may provide an effective anti-metastatic agent for CRC patients.
Collapse
Affiliation(s)
- Yiwen Jiang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Dan Hong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhefeng Lou
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuezi Tu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325000, China
| | - Longjin Jin
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
43
|
Allahverdi A, Arefian E, Soleimani M, Ai J, Nahanmoghaddam N, Yousefi-Ahmadipour A, Ebrahimi-Barough S. MicroRNA-4731-5p delivered by AD-mesenchymal stem cells induces cell cycle arrest and apoptosis in glioblastoma. J Cell Physiol 2020; 235:8167-8175. [PMID: 31957033 DOI: 10.1002/jcp.29472] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/07/2020] [Indexed: 11/10/2022]
Abstract
Glioblastoma multiforme (GBM) exhibits the most malignant brain tumor with very poor prognosis. MicroRNAs (miRNAs) are regulatory factors that can downregulate the expression of multiple genes. Several miRNAs acting as tumor-suppressor genes have been identified so far. The delivery of miRNA by mesenchymal stem cell (MSC) due to their ability to specifically target tumors is a new, hopeful therapeutic approach for glioblastoma. The objective of our study is the investigation of the effect of lentivirus-mediated microRNA-4731 (miR-4731) genetic manipulated adipose-derived (AD)-MSC on GBM. The downregulation of miR-4731 in human GBM tumor was detected using the GEO dataset. To evaluate the function of miR-4731, we overexpressed miR-4731 using lentiviral vectors in U-87 and U-251 GBM cell lines. The effects of miR-4731 on cell proliferation and cell cycle of glioma cells were analyzed by wound test and flow-cytometry assay. miR-4731 inhibited the proliferation of GBM cancer cells. Coculturing was used to study the antiproliferative effect of miR-4731-AD-MSCs on GBM cell lines. Direct and indirect coculture of GBM cell lines with miR-4731-AD-MSCs induced cell cycle arrest and apoptosis. Our findings suggest that AD-MSCs expressing miR-4731 have favorable antitumor characteristics and should be further explored in future glioma therapy.
Collapse
Affiliation(s)
- Amir Allahverdi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Molecular and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Masoud Soleimani
- Hematology and Cell therapy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Nahanmoghaddam
- Department of Pediatrics, School of Medicine, Bouali Childrens' Hospital, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Aliakbar Yousefi-Ahmadipour
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Zhang L, Wang Y, Wu G, Rao L, Wei Y, Yue H, Yuan T, Yang P, Xiong F, Zhang S, Zhou Q, Chen Z, Li J, Mo BW, Zhang H, Xiong W, Wang CY. Blockade of JAK2 protects mice against hypoxia-induced pulmonary arterial hypertension by repressing pulmonary arterial smooth muscle cell proliferation. Cell Prolif 2020; 53:e12742. [PMID: 31943454 PMCID: PMC7046303 DOI: 10.1111/cpr.12742] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 12/29/2022] Open
Abstract
Objectives Hypoxia is an important risk factor for pulmonary arterial remodelling in pulmonary arterial hypertension (PAH), and the Janus kinase 2 (JAK2) is believed to be involved in this process. In the present report, we aimed to investigate the role of JAK2 in vascular smooth muscle cells during the course of PAH. Methods Smooth muscle cell (SMC)‐specific Jak2 deficient mice and their littermate controls were subjected to normobaric normoxic or hypoxic (10% O2) challenges for 28 days to monitor the development of PAH, respectively. To further elucidate the potential mechanisms whereby JAK2 influences pulmonary vascular remodelling, a selective JAK2 inhibitor was applied to pre‐treat human pulmonary arterial smooth muscle cells (HPASMCs) for 1 hour followed by 24‐hour hypoxic exposure. Results Mice with hypoxia‐induced PAH were characterized by the altered JAK2/STAT3 activity in pulmonary artery smooth muscle cells. Therefore, induction of Jak2 deficiency in SMCs protected mice from hypoxia‐induced increase of right ventricular systolic pressure (RVSP), right ventricular hypertrophy and pulmonary vascular remodelling. Particularly, loss of Jak2 significantly attenuated chronic hypoxia‐induced PASMC proliferation in the lungs. Similarly, blockade of JAK2 by its inhibitor, TG‐101348, suppressed hypoxia‐induced human PASMC proliferation. Upon hypoxia‐induced activation, JAK2 phosphorylated signal transducer and activator of transcription 3 (STAT3), which then bound to the CCNA2 promoter to transcribe cyclin A2 expression, thereby promoting PASMC proliferation. Conclusions Our studies support that JAK2 could be a culprit contributing to the pulmonary vascular remodelling, and therefore, it could be a viable target for prevention and treatment of PAH in clinical settings.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guorao Wu
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lizong Rao
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yanqiu Wei
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huihui Yue
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yuan
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ping Yang
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Zhou
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhishui Chen
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxiu Li
- Shenzhen Third People's Hospital, Shenzhen, China
| | - Bi-Wen Mo
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Huilan Zhang
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weining Xiong
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Respiratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cong-Yi Wang
- Key Laboratory of Organ Transplantation, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Education, The Center for Biomedical Research, Chinese Academy of Medical Sciences, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Kishimoto K, Sugano-Yasunaga W, Taniguchi A, Agata K, Nonaka S, Funayama N. Skeleton construction upon local regression of the sponge body. Dev Growth Differ 2019; 61:485-500. [PMID: 31820450 DOI: 10.1111/dgd.12636] [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: 01/04/2018] [Revised: 10/04/2019] [Accepted: 10/18/2019] [Indexed: 12/09/2022]
Abstract
We previously revealed that the mechanism of demosponge skeleton construction is self-organization by multiple rounds of sequential mechanical reactions of player cells. In these reactions, "transport cells" dynamically carry fine skeletal elements (spicules) on epithelia surrounding the inner body space of sponges (basal epithelium (basopinacoderm) and the endodermal epithelium (ENCM)). Once spicules pierce ENCM and apical pinacoderm, subsequently they are cemented to the substratum under the sponge body, or connected to other skeleton-constructing spicules. Thus, the "pierce" step is the key to holding up spicules in the temporary periphery of growing sponges' bodies. Since sponges can regress as well as grow, here we asked how skeleton construction occurs during local regression of the body. We found that prior to local basopinacoderm retraction (and thus body regression), the body became thinner. Some spicules that were originally carried outward stagnated for a while, and were then carried inwards either on ENCM or basopinacoderm. Spicules that were carried inwards on ENCM pierced epithelia after a short transport, and thus became held up at relatively inward positions compared to spicules carried on outwardly extending basopinacoderm. The switch of epithelia on which transport cells migrate efficiently occurred in thinner body spaces where basopinacoderm and ENCM became close to each other. Thus, the mechanisms underlying this phenomenon are rather mechanical: the combination of sequential reactions of skeleton construction and the narrowed body space upon local retraction of basopinacoderm cause spicules to be held up at more-inward positions, which might strengthen the basopinacoderm's attachment to substratum.
Collapse
Affiliation(s)
- Kouji Kishimoto
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | | | - Atsushi Taniguchi
- Laboratory for Spatiotemporal Regulations, National Institute for Basic Biology, Okazaki, Japan
| | - Kiyokazu Agata
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan.,Laboratory of regeneration biology, National Institute for Basic Biology, Okazaki, Japan
| | - Shigenori Nonaka
- Laboratory for Spatiotemporal Regulations, National Institute for Basic Biology, Okazaki, Japan
| | - Noriko Funayama
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| |
Collapse
|
46
|
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: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
47
|
Qin X, Chen R, Xiong R, Tan Z, Gao S, Lin C, Huo T. Comprehensive analysis of non-small-cell lung cancer microarray datasets identifies several prognostic biomarkers. Future Oncol 2019; 15:3135-3148. [PMID: 31426680 DOI: 10.2217/fon-2018-0824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To find accurate and effective biomarkers for diagnosis of non-small-cell lung cancer (NSCLC) patients. Materials & methods: We downloaded microarray datasets GSE19188, GSE33532, GSE101929 and GSE102286 from the database of Gene Expression Omnibus. We screened out differentially expressed genes (DEGs) and miRNAs (DEMs) with GEO2R. We also performed analyses for the enrichment of DEGs' and DEMs' function and pathway by several tools including database for annotation, visualization and integrated discovery, protein-protein interaction and Kaplan-Meier-plotter. Results: Total 913 DEGs were screened out, among which ten hub genes were discovered. All the hub genes were linked to the worsening overall survival of the NSCLC patients. Besides, 98 DEMs were screened out. MiR-9 and miR-520e were the most significantly regulated miRNAs. Conclusion: Our results could provide potential targets for the diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Xiuxiu Qin
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, PR China
| | - Ruoshi Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, PR China
| | - Rui Xiong
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, PR China
| | - Zimiao Tan
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, PR China
| | - Shanshan Gao
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, PR China
| | - Chunshui Lin
- Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, PR China
| | - Tianming Huo
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, PR China
| |
Collapse
|
48
|
Jones MC, Zha J, Humphries MJ. Connections between the cell cycle, cell adhesion and the cytoskeleton. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180227. [PMID: 31431178 PMCID: PMC6627016 DOI: 10.1098/rstb.2018.0227] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Cell division, the purpose of which is to enable cell replication, and in particular to distribute complete, accurate copies of genetic material to daughter cells, is essential for the propagation of life. At a morphological level, division not only necessitates duplication of cellular structures, but it also relies on polar segregation of this material followed by physical scission of the parent cell. For these fundamental changes in cell shape and positioning to be achieved, mechanisms are required to link the cell cycle to the modulation of cytoarchitecture. Outside of mitosis, the three main cytoskeletal networks not only endow cells with a physical cytoplasmic skeleton, but they also provide a mechanism for spatio-temporal sensing via integrin-associated adhesion complexes and site-directed delivery of cargoes. During mitosis, some interphase functions are retained, but the architecture of the cytoskeleton changes dramatically, and there is a need to generate a mitotic spindle for chromosome segregation. An economical solution is to re-use existing cytoskeletal molecules: transcellular actin stress fibres remodel to create a rigid cortex and a cytokinetic furrow, while unipolar radial microtubules become the primary components of the bipolar spindle. This remodelling implies the existence of specific mechanisms that link the cell-cycle machinery to the control of adhesion and the cytoskeleton. In this article, we review the intimate three-way connection between microenvironmental sensing, adhesion signalling and cell proliferation, particularly in the contexts of normal growth control and aberrant tumour progression. As the morphological changes that occur during mitosis are ancient, the mechanisms linking the cell cycle to the cytoskeleton/adhesion signalling network are likely to be primordial in nature and we discuss recent advances that have elucidated elements of this link. A particular focus is the connection between CDK1 and cell adhesion. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
| | | | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
49
|
Liu F, Wu Y, Mi Y, Gu L, Sang M, Geng C. Identification of core genes and potential molecular mechanisms in breast cancer using bioinformatics analysis. Pathol Res Pract 2019; 215:152436. [DOI: 10.1016/j.prp.2019.152436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/11/2019] [Accepted: 05/03/2019] [Indexed: 12/11/2022]
|
50
|
Lv X, He M, Zhao Y, Zhang L, Zhu W, Jiang L, Yan Y, Fan Y, Zhao H, Zhou S, Ma H, Sun Y, Li X, Xu H, Wei M. Identification of potential key genes and pathways predicting pathogenesis and prognosis for triple-negative breast cancer. Cancer Cell Int 2019; 19:172. [PMID: 31297036 PMCID: PMC6599314 DOI: 10.1186/s12935-019-0884-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Background Triple negative breast cancer (TNBC) is a specific subtype of breast cancer with a poor prognosis due to its aggressive biological behaviour and lack of therapeutic targets. We aimed to explore some novel genes and pathways related to TNBC prognosis through bioinformatics methods as well as potential initiation and progression mechanisms. Methods Breast cancer mRNA data were obtained from The Cancer Genome Atlas database (TCGA). Differential expression analysis of cancer and adjacent cancer, as well as, triple negative breast cancer and non-triple negative breast cancer were performed using R software. The key genes related to the pathogenesis were identified by functional and pathway enrichment analysis and protein-protein interaction network analysis. Based on univariate and multivariate Cox proportional hazards model analyses, a gene signature was established to predict overall survival. Receiver operating characteristic curve was used to evaluate the prognostic performance of our model. Results Based on mRNA expression profiling of breast cancer patients from the TCGA database, 755 differentially expressed overlapping mRNAs were detected between TNBC/non-TNBC samples and normal tissue. We found eight hub genes associated with the cell cycle pathway highly expressed in TNBC. Additionally, a novel six-gene (TMEM252, PRB2, SMCO1, IVL, SMR3B and COL9A3) signature from the 755 differentially expressed mRNAs was constructed and significantly associated with prognosis as an independent prognostic signature. TNBC patients with high-risk scores based on the expression of the 6-mRNAs had significantly shorter survival times compared to patients with low-risk scores (P < 0.0001). Conclusions The eight hub genes we identified might be tightly correlated with TNBC pathogenesis. The 6-mRNA signature established might act as an independent biomarker with a potentially good performance in predicting overall survival.
Collapse
Affiliation(s)
- Xuemei Lv
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Miao He
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yanyun Zhao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Liwen Zhang
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Wenjing Zhu
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Longyang Jiang
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yuanyuan Yan
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yue Fan
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Hongliang Zhao
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Shuqi Zhou
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Heyao Ma
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Yezhi Sun
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| | - Xiang Li
- 3Department of Breast Cancer, Cancer Hospital of China Medical University, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042 China
| | - Hong Xu
- 3Department of Breast Cancer, Cancer Hospital of China Medical University, No. 44, Xiaoheyan Road, Dadong District, Shenyang, 110042 China
| | - Minjie Wei
- 1Department of Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-tumour Drug Development and Evaluation, Shenyang, China
| |
Collapse
|