1
|
Liu L, Wu M, Chen Y, Cheng Y, Liu S, Zhang X, Xie Q, Cao L, Wei L, Fang Y, Jafri A, Sferra TJ, Shen A, Li L. Downregulating FGGY carbohydrate kinase domain containing promotes cell senescence by activating the p53/p21 signaling pathway in colorectal cancer. Int J Mol Med 2025; 55:81. [PMID: 40116125 PMCID: PMC11964412 DOI: 10.3892/ijmm.2025.5522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/14/2025] [Indexed: 03/23/2025] Open
Abstract
Carbohydrate kinases serve an oncogenic role in several types of cancer; however, the function of FGGY carbohydrate kinase domain containing (FGGY) in colorectal cancer (CRC) remains unknown. The present study investigated the function and possible molecular mechanisms of FGGY in CRC. The results showed that elevated levels of FGGY mRNA and protein were observed in CRC tissues, and a higher expression of FGGY was associated with advanced N stage and reduced overall survival time in patients with CRC. Silencing FGGY inhibited the viability of CRC cells by inducing cell cycle arrest and promoting apoptosis in vitro, thereby attenuating tumor growth in a xenograft mouse model. FGGY knockdown also enriched the senescence‑associated heterochromatin foci (SAHF) pathway and p53 pathway, as further confirmed by enhancing senescence‑associated β‑galactosidase (SA‑β‑gal) activity, with increased levels of SAHF‑associated proteins HP1γ and trimethylation of H3K9 (H3k9me3) in CRC cells, as well as upregulation of p53 and its downstream protein p21. Furthermore, p53 knockout rescued FGGY knockdown‑mediated reductions in cell viability, SA‑β‑gal activity, and the levels of HP1γ and H3k9me3 in CRC cells. These findings indicated that FGGY could act as a newly identified potential oncogene in CRC, partially through regulating the p53/p21 signaling pathway and altering cell senescence.
Collapse
Affiliation(s)
- Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Department of Scientific Research, Affiliated Sanming Integrated Medicine Hospital of Fujian University of Traditional Chinese Medicine, Sanming, Fujian 365001, P.R. China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Sijia Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xinran Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Liujing Cao
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Yi Fang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Anjum Jafri
- Department of Genetics and Genome Sciences, Histology Core, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas J. Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Li Li
- Shengli Clinical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
- Department of Health Management, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
2
|
Hong J, Sui P, Li Y, Xu KY, Lee JH, Wang J, Chen S, Zhang P, Wingate N, Noor A, Yuan Y, Hromas R, Zhou H, Hamamoto K, Su R, Yin CC, Ye F, Quesada AE, Chen J, Huang S, Zhou D, You MJ, Yang FC, Wang J, Xu M. PSPC1 exerts an oncogenic role in AML by regulating a leukemic transcription program in cooperation with PU.1. Cell Stem Cell 2025; 32:463-478.e6. [PMID: 39954676 DOI: 10.1016/j.stem.2025.01.010] [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: 12/28/2023] [Revised: 11/01/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy characterized by the blockage of myeloid cell differentiation and uncontrolled proliferation of immature myeloid cells. Here, we show that paraspeckle component 1 (PSPC1) is aberrantly overexpressed and associated with poor survival in AML patients. Using human AML cells and mouse models, we demonstrate that PSPC1 is not required for normal hematopoiesis, but it is critical and essential for AML cells to maintain their leukemic characteristics. PSPC1 loss induces robust differentiation, suppresses proliferation, and abolishes leukemogenesis in diverse AML cells. Mechanistically, PSPC1 exerts a pro-leukemia effect by regulating a unique leukemic transcription program via cooperative chromatin binding with PU.1 and activation of tumor-promoting genes, including NDC1, which is not previously implicated in AML. Our findings uncover a unique and crucial role of PSPC1 dependency in AML and highlight its potential as a promising therapeutic target for AML.
Collapse
Affiliation(s)
- Juyeong Hong
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pinpin Sui
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ying Li
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kerryn Y Xu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ji-Hoon Lee
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Juan Wang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Shi Chen
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Peng Zhang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Noah Wingate
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Asra Noor
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Yaxia Yuan
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Robert Hromas
- Department of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Hongwei Zhou
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karina Hamamoto
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rui Su
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - C Cameron Yin
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Fengxi Ye
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Andrés E Quesada
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Chen
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA 91016, USA
| | - Suming Huang
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - M James You
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Feng-Chun Yang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | - Jianlong Wang
- Department of Medicine, Columbia Center for Human Development and Stem Cell Therapies, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Mingjiang Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
3
|
Shodry S, Hasan YTN, Ahdi IR, Ulhaq ZS. Gene targets with therapeutic potential in hepatocellular carcinoma. World J Gastrointest Oncol 2024; 16:4543-4547. [PMID: 39678796 PMCID: PMC11577361 DOI: 10.4251/wjgo.v16.i12.4543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/03/2024] [Accepted: 08/13/2024] [Indexed: 11/12/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide. Major treatments include liver transplantation, resection, and chemotherapy, but the 5-year recurrence rate remains high. Late diagnosis often prevents surgical intervention, contributing to poor patient survival rates. Carcinogenesis in HCC involves genetic alterations that drive the transformation of normal cells into malignant ones. Enhancer of zeste homolog 2 (EZH2), a key regulator of cell cycle progression, is frequently upregulated in HCC and is associated with advanced stages and poor prognosis, making it a potential biomarker. Additionally, signal transducer and activator of transcription 3, which binds to EZH2, affects disease staging and outcomes. Targeting EZH2 presents a promising therapeutic strategy. On the other hand, abnormal lipid metabolism is a hallmark of HCC and impacts prognosis. Fatty acid binding protein 5 is highly expressed in HCC tissues and correlates with key oncogenes, suggesting its potential as a biomarker. Other genes such as guanine monophosphate synthase, cell division cycle associated 5, and epidermal growth factor receptor provide insights into the molecular mechanisms of HCC, offering potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Syifaus Shodry
- Faculty of Medicine and Health Sciences, Maulana Ibrahim Islamic State University of Malang, Malang 65144, Jawa Timur, Indonesia
| | - Yuliono Trika Nur Hasan
- Faculty of Medicine and Health Sciences, Maulana Ibrahim Islamic State University of Malang, Malang 65144, Jawa Timur, Indonesia
| | - Iwal Reza Ahdi
- Faculty of Medicine and Health Sciences, Maulana Ibrahim Islamic State University of Malang, Malang 65144, Jawa Timur, Indonesia
| | - Zulvikar Syambani Ulhaq
- Research Center for Preclinical and Clinical Medicine, National Research and Innovation Agency Republic of Indonesia, Cibinong 16911, Indonesia
| |
Collapse
|
4
|
Wang Z, Ren M, Liu W, Wu J, Tang P. Role of cell division cycle-associated proteins in regulating cell cycle and promoting tumor progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189147. [PMID: 38955314 DOI: 10.1016/j.bbcan.2024.189147] [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: 12/19/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
The cell division cycle-associated protein (CDCA) family is important in regulating cell division. High CDCA expression is significantly linked to tumor development. This review summarizes clinical and basic studies on CDCAs conducted in recent decades. Furthermore, it systematically introduces the molecular expression and function, key mechanisms, cell cycle regulation, and roles of CDCAs in tumor development, cell proliferation, drug resistance, invasion, and metastasis. Additionally, it presents the latest research on tumor diagnosis, prognosis, and treatment targeting CDCAs. These findings are pivotal for further in-depth studies on the role of CDCAs in promoting tumor development and provide theoretical support for their application as new anti-tumor targets.
Collapse
Affiliation(s)
- Zhaoyu Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China
| | - Minshijing Ren
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China
| | - Wei Liu
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China
| | - Jin Wu
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China; Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| | - Peng Tang
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China.
| |
Collapse
|
5
|
Wang X, Shi A, Liu J, Kong W, Huang Y, Xue W, Yang F, Huang J. CDCA5-EEF1A1 interaction promotes progression of clear cell renal cell carcinoma by regulating mTOR signaling. Cancer Cell Int 2024; 24:147. [PMID: 38658931 PMCID: PMC11044369 DOI: 10.1186/s12935-024-03330-4] [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: 07/30/2023] [Accepted: 04/18/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Cell division cycle associated 5 (CDCA5) plays ontogenetic role in various human cancers. However, its specific function and regulatory mechanism in ccRCC remain uncertain. METHODS Immunohistochemistry and western blots were performed to investigate the expression of CDCA5 in ccRCC tissues. Genetic knockdown and upregulation of CDCA5 were performed to investigate its functional roles in ccRCC proliferation, migration, apoptosis and sunitinib resistance. Furthermore, Co-IP assay and LC-MS/MS were performed to investigate the underlying mechanisms. RESULTS We found that CDCA5 expression is frequently upregulated in ccRCC tumors and is associated with poor prognosis of ccRCC patients. Functionally, CDCA5 promotes proliferation, migration, and sunitinib resistance, while inhibiting apoptosis in ccRCC cells. In vivo mouse xenograft model confirms that silencing of CDCA5 drastically inhibits the growth of ccRCC. Mechanistically, we discovered that CDCA5 interacts with Eukaryotic Translation Elongation Factor 1 Alpha 1 (EEF1A1) to regulate mTOR signaling pathway, thereby promoting ccRCC progression. CONCLUSIONS Taken together, our results demonstrate the significant role of CDCA5 in ccRCC progression. The findings may provide insights for the development of new treatment strategies targeting CDCA5 for ccRCC patients.
Collapse
Affiliation(s)
- Xun Wang
- Department of Urology, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - An Shi
- Department of Critical Care Medicine, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Jie Liu
- Department of Pharmacy, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wen Kong
- Department of Urology, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Yiran Huang
- Department of Urology, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Fan Yang
- Department of Pharmacy, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jiwei Huang
- Department of Urology, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
6
|
de Castro JNP, da Silva Costa SM, Camargo ACL, Ito MT, de Souza BB, de Haidar E Bertozzo V, Rodrigues TAR, Lanaro C, de Albuquerque DM, Saez RC, Saad STO, Ozelo MC, Cendes F, Costa FF, de Melo MB. Comparative transcriptomic analysis of circulating endothelial cells in sickle cell stroke. Ann Hematol 2024; 103:1167-1179. [PMID: 38386032 DOI: 10.1007/s00277-024-05655-6] [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: 10/11/2023] [Accepted: 02/08/2024] [Indexed: 02/23/2024]
Abstract
Ischemic stroke (IS) is one of the most impairing complications of sickle cell anemia (SCA), responsible for 20% of mortality in patients. Rheological alterations, adhesive properties of sickle reticulocytes, leukocyte adhesion, inflammation and endothelial dysfunction are related to the vasculopathy observed prior to ischemic events. The role of the vascular endothelium in this complex cascade of mechanisms is emphasized, as well as in the process of ischemia-induced repair and neovascularization. The aim of the present study was to perform a comparative transcriptomic analysis of endothelial colony-forming cells (ECFCs) from SCA patients with and without IS. Next, to gain further insights of the biological relevance of differentially expressed genes (DEGs), functional enrichment analysis, protein-protein interaction network (PPI) construction and in silico prediction of regulatory factors were performed. Among the 2469 DEGs, genes related to cell proliferation (AKT1, E2F1, CDCA5, EGFL7), migration (AKT1, HRAS), angiogenesis (AKT1, EGFL7) and defense response pathways (HRAS, IRF3, TGFB1), important endothelial cell molecular mechanisms in post ischemia repair were identified. Despite the severity of IS in SCA, widely accepted molecular targets are still lacking, especially related to stroke outcome. The comparative analysis of the gene expression profile of ECFCs from IS patients versus controls seems to indicate that there is a persistent angiogenic process even after a long time this complication has occurred. Thus, this is an original study which may lead to new insights into the molecular basis of SCA stroke and contribute to a better understanding of the role of endothelial cells in stroke recovery.
Collapse
Affiliation(s)
- Júlia Nicoliello Pereira de Castro
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Sueli Matilde da Silva Costa
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Ana Carolina Lima Camargo
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Mirta Tomie Ito
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Bruno Batista de Souza
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Victor de Haidar E Bertozzo
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Thiago Adalton Rosa Rodrigues
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil
| | - Carolina Lanaro
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | | | - Roberta Casagrande Saez
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Margareth Castro Ozelo
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Fernando Cendes
- Neuroimaging Laboratory, Department of Neurology, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - Mônica Barbosa de Melo
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering-CBMEG, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo, 13083-875, Brazil.
| |
Collapse
|
7
|
Chen X, Zhou M, Ma S, Wu H, Cai H. KLF5-mediated CDCA5 expression promotes tumor development and progression of epithelial ovarian carcinoma. Exp Cell Res 2023; 429:113645. [PMID: 37247719 DOI: 10.1016/j.yexcr.2023.113645] [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: 11/22/2022] [Revised: 05/10/2023] [Accepted: 05/13/2023] [Indexed: 05/31/2023]
Abstract
Cell division cycle associated 5 (CDCA5) is correlated with the development and progression of many malignant tumors. However, little is known about its role in epithelial ovarian cancer (EOC) progression. In this study, the clinical value, biological function and underlying mechanisms of CDCA5 in EOC were evaluated. CDCA5 mRNA and protein levels were substantially upregulated in EOC and had a significant positive correlation with adverse clinicopathological characteristics and a poor prognosis. CDCA5 facilitated proliferation, invasion, and metastasis and disrupted mitochondrial-mediated endogenous apoptosis by activating the cell cycle pathway and inhibiting the P53 pathway in EOC cells. Conversely, knockdown of CDCA5 expression blocked the malignant activities of EOC cells and suppressed the growth of xenograft tumors in vivo. Mechanistically, the transcription factor KLF5 bound to a specific site in the CDCA5 promoter and promoted CDCA5 expression. Moreover, KLF5 overexpression rescued the negative regulation of inhibited CDCA5 expression on EOC cell proliferation. In conclusion, our findings revealed that CDCA5 promoted tumor progression of EOC via the KLF5/CDCA5/cell cycle and P53 axes, which might provide new insights into the roles of CDCA5 in EOC.
Collapse
Affiliation(s)
- Xiaohong Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730030, China; Department of Gynecology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Meiying Zhou
- Department of Gynecology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Shouye Ma
- Department of Gynecology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Huifang Wu
- Department of Gynecology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hui Cai
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730030, China; Department of Surgery, Gansu Provincial Hospital, Lanzhou, 730000, China.
| |
Collapse
|
8
|
Song Y, Huang T, Pan H, Du A, Wu T, Lan J, Zhou X, Lv Y, Xue S, Yuan K. The influence of COVID-19 on colorectal cancer was investigated using bioinformatics and systems biology techniques. Front Med (Lausanne) 2023; 10:1169562. [PMID: 37457582 PMCID: PMC10348756 DOI: 10.3389/fmed.2023.1169562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) is a global pandemic and highly contagious, posing a serious threat to human health. Colorectal cancer (CRC) is a risk factor for COVID-19 infection. Therefore, it is vital to investigate the intrinsic link between these two diseases. Methods In this work, bioinformatics and systems biology techniques were used to detect the mutual pathways, molecular biomarkers, and potential drugs between COVID-19 and CRC. Results A total of 161 common differentially expressed genes (DEGs) were identified based on the RNA sequencing datasets of the two diseases. Functional analysis was performed using ontology keywords, and pathway analysis was also performed. The common DEGs were further utilized to create a protein-protein interaction (PPI) network and to identify hub genes and key modules. The datasets revealed transcription factors-gene interactions, co-regulatory networks with DEGs-miRNAs of common DEGs, and predicted possible drugs as well. The ten predicted drugs include troglitazone, estradiol, progesterone, calcitriol, genistein, dexamethasone, lucanthone, resveratrol, retinoic acid, phorbol 12-myristate 13-acetate, some of which have been investigated as potential CRC and COVID-19 therapies. Discussion By clarifying the relationship between COVID-19 and CRC, we hope to provide novel clues and promising therapeutic drugs to treat these two illnesses.
Collapse
Affiliation(s)
- Yujia Song
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyuan Pan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ao Du
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Wu
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jiang Lan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Zhou
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Lv
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuai Xue
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Gao Y, Liu S, Yang J, Su M, Xu J, Wang H, Zhang J. The Comprehensive Analysis Illustrates the Role of CDCA5 in Breast Cancer: An Effective Diagnosis and Prognosis Biomarker. Int J Genomics 2023; 2023:7150141. [PMID: 37287817 PMCID: PMC10243952 DOI: 10.1155/2023/7150141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/26/2023] [Accepted: 05/10/2023] [Indexed: 06/09/2023] Open
Abstract
Background Several studies have been conducted to investigate the role of cell division cycle-associated 5 (CDCA5) in cancer. Its role in breast cancer, however, remains unknown. Methods The Gene Expression Omnibus and Cancer Genome Atlas Program databases provided the open-access information needed for the research. The CCK8 and colony formation assays were used to measure cell proliferation. The capacity of breast cancer cells to invade and migrate was assessed using the transwell assay. Results In our study, CDCA5 was identified as the interested gene through a series of bioinformatics analysis. We found a higher CDCA5 expression level in tissue and cells of breast cancer. Meanwhile, CDCA5 has been linked to increased proliferation, invasion, and migration of breast cancer cells, which was also associated with worse clinical features. The biochemical pathways, in which CDCA5 was engaged, were identified using biological enrichment analysis. Immune infiltration research revealed that CDCA5 was linked to enhanced activity of several immune function terms. Meanwhile, DNA methylation might be responsible for the aberrant level of CDCA5 in tumor tissue. In addition, CDCA5 could significantly increase the paclitaxel and docetaxel sensitivity, indicating that it has the potential for clinical application. Also, we found that CDCA5 is mainly localized in cell nucleoplasm. Moreover, in the breast cancer microenvironment, we found that CDCA5 is mainly expressed in malignant cells, proliferation T cells, and neutrophils. Conclusion Overall, our findings suggest that CDCA5 is a potential prognostic indicator and target for breast cancer, which can indicate the direction of the relevant research.
Collapse
Affiliation(s)
- Yang Gao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Shuting Liu
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- Department of Breast and Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Junyuan Yang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Min Su
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Jingjing Xu
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- Department of Breast and Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hua Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Jingwei Zhang
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- Department of Breast and Thyroid Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
10
|
Di X, Xiang L, Jian Z. YAP-mediated mechanotransduction in urinary bladder remodeling: Based on RNA-seq and CUT&Tag. Front Genet 2023; 14:1106927. [PMID: 36741311 PMCID: PMC9895788 DOI: 10.3389/fgene.2023.1106927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
Yes-associated protein (YAP) is an important transcriptional coactivator binding to transcriptional factors that engage in many downstream gene transcription. Partial bladder outlet obstruction (pBOO) causes a massive burden to patients and finally leads to bladder fibrosis. Several cell types engage in the pBOO pathological process, including urothelial cells, smooth muscle cells, and fibroblasts. To clarify the function of YAP in bladder fibrosis, we performed the RNA-seq and CUT&Tag of the bladder smooth muscle cell to analyze the YAP ablation of human bladder smooth muscle cells (hBdSMCs) and immunoprecipitation of YAP. 141 differentially expressed genes (DEGs) were identified through RNA-seq between YAP-knockdown and nature control. After matching with the results of CUT&Tag, 36 genes were regulated directly by YAP. Then we identified the hub genes in the DEGs, including CDCA5, CENPA, DTL, NCAPH, and NEIL3, that contribute to cell proliferation. Thus, our study provides a regulatory network of YAP in smooth muscle proliferation. The possible effects of YAP on hBdSMC might be a vital target for pBOO-associated bladder fibrosis.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liyuan Xiang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongyu Jian
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Zhongyu Jian,
| |
Collapse
|
11
|
Multiomics Study of a Novel Naturally Derived Small Molecule, NSC772864, as a Potential Inhibitor of Proto-Oncogenes Regulating Cell Cycle Progression in Colorectal Cancer. Cells 2023; 12:cells12020340. [PMID: 36672275 PMCID: PMC9856482 DOI: 10.3390/cells12020340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant tumors, and it contributes to high numbers of deaths globally. Although advances in understanding CRC molecular mechanisms have shed significant light on its pathogenicity, current treatment options, including combined chemotherapy and molecular-targeted agents, are still limited due to resistance, with almost 25% of patients developing distant metastasis. Therefore, identifying novel biomarkers for early diagnosis is crucial, as they will also influence strategies for new targeted therapies. The proto-oncogene, c-Met, a tyrosine kinase that promotes cell proliferation, motility, and invasion; c-MYC, a transcription factor associated with the modulation of the cell cycle, proliferation, apoptosis; and cyclin D1 (CCND1), an essential regulatory protein in the cell cycle, all play crucial roles in cancer progression. In the present study, we explored computational simulations through bioinformatics analysis and identified the overexpression of c-Met/GSK3β/MYC/CCND1 oncogenic signatures that were associated with cancer progression, drug resistance, metastasis, and poor clinical outcomes in CRC. We further demonstrated the anticancer activities of our newly synthesized quinoline-derived compound, NSC772864, against panels of the National Cancer Institute's human CRC cell lines. The compound exhibited cytotoxic activities against various CRC cell lines. Using target prediction tools, we found that c-Met/GSK3β/MYC/CCND1 were target genes for the NSC772864 compound. Subsequently, we performed in silico molecular docking to investigate protein-ligand interactions and discovered that NSC772864 exhibited higher binding affinities with these oncogenes compared to FDA-approved drugs. These findings strongly suggest that NSC772864 is a novel and potential antiCRC agent.
Collapse
|
12
|
Xu J, Wang J, Zhao M, Li C, Hong S, Zhang J. LncRNA LINC01018/miR-942-5p/KNG1 axis regulates the malignant development of glioma in vitro and in vivo. CNS Neurosci Ther 2022; 29:691-711. [PMID: 36550594 PMCID: PMC9873518 DOI: 10.1111/cns.14053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
AIMS Since the inhibitory effect of KNG1 on glioma has been proved, this study further explores the regulation of the lncRNA/miRNA axis on KNG1 in glioma. METHODS The miRNAs that target KNG1 and the lncRNA that targets miR-942-5p were predicted by bioinformatics analysis and verified by experiments. The correlations between miR-942-5p and the survival of patients and between KNG1 and miR-942-5p were analyzed. After transfection, cell migration, invasion, proliferation, and cell cycle were detected through wound healing, Transwell, colony formation, and flow cytometry assays. A mouse subcutaneous xenotransplanted tumor model was established. The expressions of miR-942-5p, KNG1, LINC01018, and related genes were evaluated by quantitative real-time reverse transcription polymerase chain reaction (RT-qPCR), Western blot, or immunohistochemistry. RESULTS MiR-942-5p targeted KNG1, and LINC01018 sponged miR-942-5p. The high survival rate of patients was related to low miR-942-5p level. MiR-942-5p was highly expressed, whereas KNG1 was lowly expressed in glioma. MiR-942-5p was negatively correlated with KNG1. Silent LINC01018 or KNG1 and miR-942-5p mimic enhanced the migration, invasion, and proliferation of glioma cells, and regulated the expressions of metastasis-related and proliferation-related genes. LINC01018 knockdown and miR-942-5p mimic promoted glioma tumor growth in mice. The levels of miR-942-5p and KNG1 were decreased by LINC01018 knockdown, and LINC01018 expression was suppressed by miR-942-5p mimic. MiR-942-5p inhibitor, KNG1, and LINC01018 had the opposite effect to miR-942-5p mimic. CONCLUSION LINC01018/miR-942-5p/KNG1 pathway regulates the development of glioma cells in vitro and in vivo.
Collapse
Affiliation(s)
- Jinfang Xu
- Department of NeurosurgeryThe Second Affiliated Hospital Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Jianli Wang
- Department of NeurosurgeryThe Second Affiliated Hospital Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Mingfei Zhao
- Department of NeurosurgeryThe Second Affiliated Hospital Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Chenguang Li
- Department of NeurosurgeryThe Second Affiliated Hospital Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Shen Hong
- Department of NeurosurgeryThe Second Affiliated Hospital Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Jianmin Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital Zhejiang University School of MedicineHangzhouZhejiangChina
| |
Collapse
|
13
|
Li X, Abdel-Maksoud MA, Iqbal I, Mubarak A, Farrag MA, Haris M, Alghamdi S, Ain QU, Almekhlafi S. Deciphering cervical cancer-associated biomarkers by integrated multi-omics approach. Am J Transl Res 2022; 14:8843-8861. [PMID: 36628250 PMCID: PMC9827308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/13/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVES Cervical Squamous Cell Carcinoma (CESC) is one of the most fatal female malignancies, and the underlying molecular mechanisms governing this disease have not been fully explored. In this research, we planned to conduct the analysis of Gene Expression Omnibus (GEO) cervical squamous cell carcinoma microarray datasets by a detailed in silico approach and to explore some novel biomarkers of CESC. METHODS The top commonly differentially expressed genes (DEGs) from the GSE138080 and GSE113942 datasets were analyzed by Limma package-based GEO2R tool. The protein-protein interaction (PPI) network of the DEGs was drawn through Search Tool for the Retrieval of Interacting Genes (STRING), and top 6 hub genes were obtained from Cytoscape. Expression analysis and validation of hub genes expression in CESC samples and cell lines were done using UALCAN, OncoDB, GENT2, and HPA. Additionally, cBioPortal, Gene set enrichment analysis (GSEA) tool, Kaplan-Meier (KM) plotter, ShinyGO, and DGIdb databases were also used to check some important values of hub genes in CESC. RESULTS Out of 79 DEGs, the minichromosome maintenance complex component 4 (MCM4), nucleolar and spindle-associated protein 1 (NUSAP1), cell division cycle associated 5 (CDCA5), cell division cycle 45 (CDC45), denticleless E3 ubiquitin protein ligase homolog (DTL), and chromatin licensing and DNA replication factor 1 (CDT1) genes were regarded as hub genes in CESC. Further analysis revealed that the expressions of all these hub genes were significantly elevated in CESC cell lines and samples of diverse clinical attributes. In this study, we also documented some important correlations between hub genes and some other diverse measures, including DNA methylation, genetic alterations, and Overall Survival (OS). Last, we also identify hub genes associated ceRNA network and 31 important chemotherapeutic drugs. CONCLUSION Through detailed in silico methodology, we identified 6 hub genes, including MCM4, NUSAP1, CDCA5, CDC45, DTL, and CDT1, which are likely to be associated with CESC development and diagnosis.
Collapse
Affiliation(s)
- Xuhong Li
- Department of Gynaecology and Obstetrics, Shanghai Eighth People’s HospitalShanghai, China
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Iqra Iqbal
- Azra Naheed Medical CollegeLahore, Pakistan
| | - Ayman Mubarak
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Mohamed A Farrag
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Muhammad Haris
- Department of Anatomy, Institute of Basic Medical Sciences, Khyber Medical UniversityPeshawar, Pakistan
| | - Sumaiah Alghamdi
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| | - Qurat Ul Ain
- Anhui Provincial Hospital, Division of Life Science and Medicine, University of Science and Technology ChinaHefei, Anhui, China
| | - Sally Almekhlafi
- Department of Botany and Microbiology, College of Science, King Saud UniversityP.O. 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
14
|
Kariri YA, Joseph C, Alsaleem MA, Elsharawy KA, Alsaeed S, Toss MS, Mongan NP, Green AR, Rakha EA. Mechanistic and Clinical Evidence Supports a Key Role for Cell Division Cycle Associated 5 (CDCA5) as an Independent Predictor of Outcome in Invasive Breast Cancer. Cancers (Basel) 2022; 14:cancers14225643. [PMID: 36428736 PMCID: PMC9688237 DOI: 10.3390/cancers14225643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cell Division Cycle Associated 5 (CDCA5) plays a role in the phosphoinositide 3-kinase (PI3K)/AKT/mTOR signalling pathway involving cell division, cancer cell migration and apoptosis. This study aims to assess the prognostic and biological value of CDCA5 in breast cancer (BC). METHODS The biological and prognostic value of CDCA5 were evaluated at mRNA (n = 5109) and protein levels (n = 614) utilizing multiple well-characterized early stage BC cohorts. The effects of CDCA5 knockdown (KD) on multiple oncogenic assays were assessed in vitro using a panel of BC cell lines. RESULTS this study examined cohorts showed that high CDCA5 expression was correlated with features characteristic of aggressive behavior and poor prognosis, including the presence of high grade, large tumor size, lymphovascular invasion (LVI), hormone receptor negativity and HER2 positivity. High CDCA5 expression, at both mRNA and protein levels, was associated with shorter BC-specific survival independent of other variables (p = 0.034, Hazard ratio (HR) = 1.6, 95% CI; 1.1-2.3). In line with the clinical data, in vitro models indicated that CDCA5 depletion results in a marked decrease in BC cell invasion and migration abilities and a significant accumulation of the BC cells in the G2/M-phase. CONCLUSIONS These results provide evidence that CDCA5 plays an important role in BC development and metastasis and could be used as a potential biomarker to predict disease progression in BC.
Collapse
Affiliation(s)
- Yousif A. Kariri
- Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University Park Campus, University of Nottingham, Nottingham NG7 2RD, UK
- Department of Clinical Laboratory Science, Faculty of Applied Medical Science, Shaqra University, Shaqra 11961, Saudi Arabia
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
| | - Chitra Joseph
- School of Medicine, Nottingham City Hospital, Nottingham University Hospitals NHS Trust and The University of Nottingham, Nottingham NG5 1PB, UK
| | - Mansour A. Alsaleem
- Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University Park Campus, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
- Department of Applied Medical Science, Applied College, Qassim University, Unayzah 56435, Saudi Arabia
| | - Khloud A. Elsharawy
- Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University Park Campus, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
- Department of Zoology, Faculty of Science, Damietta University, Damietta 34517, Egypt
| | - Sami Alsaeed
- Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University Park Campus, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
- Department of Clinical Laboratory Science, Faculty of Applied Medical Sciences, Northern Border University, Arar 73244, Saudi Arabia
| | - Michael S. Toss
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
- School of Medicine, Nottingham City Hospital, Nottingham University Hospitals NHS Trust and The University of Nottingham, Nottingham NG5 1PB, UK
| | - Nigel P. Mongan
- Biodiscovery Institute, Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham NG7 2RD, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrew R. Green
- Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University Park Campus, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
| | - Emad A. Rakha
- Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University Park Campus, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Nottingham NG7 2RD, UK
- School of Medicine, Nottingham City Hospital, Nottingham University Hospitals NHS Trust and The University of Nottingham, Nottingham NG5 1PB, UK
- Correspondence: or ; Tel.: +44-0115-9691169; Fax: +44-0115-9627768
| |
Collapse
|
15
|
He J, Zhou X, Wang X, Zhang Q, Zhang L, Wang T, Zhu W, Liu P, Zhu M. Prognostic and Immunological Roles of Cell Cycle Regulator CDCA5 in Human Solid Tumors. Int J Gen Med 2022; 15:8257-8274. [DOI: 10.2147/ijgm.s389275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/02/2022] [Indexed: 11/22/2022] Open
|
16
|
Deng Y, Huang H, Shi J, Jin H. Identification of Candidate Genes in Breast Cancer Induced by Estrogen Plus Progestogens Using Bioinformatic Analysis. Int J Mol Sci 2022; 23:ijms231911892. [PMID: 36233194 PMCID: PMC9569986 DOI: 10.3390/ijms231911892] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Menopausal hormone therapy (MHT) was widely used to treat menopause-related symptoms in menopausal women. However, MHT therapies were controversial with the increased risk of breast cancer because of different estrogen and progestogen combinations, and the molecular basis behind this phenomenon is currently not understood. To address this issue, we identified differentially expressed genes (DEGs) between the estrogen plus progestogens treatment (EPT) and estrogen treatment (ET) using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) data. As a result, a total of 96 upregulated DEGs were first identified. Seven DEGs related to the cell cycle (CCNE2, CDCA5, RAD51, TCF19, KNTC1, MCM10, and NEIL3) were validated by RT-qPCR. Specifically, these seven DEGs were increased in EPT compared to ET (p < 0.05) and had higher expression levels in breast cancer than adjacent normal tissues (p < 0.05). Next, we found that estrogen receptor (ER)-positive breast cancer patients with a higher CNNE2 expression have a shorter overall survival time (p < 0.05), while this effect was not observed in the other six DEGs (p > 0.05). Interestingly, the molecular docking results showed that CCNE2 might bind to 17β-estradiol (−6.791 kcal/mol), progesterone (−6.847 kcal/mol), and medroxyprogesterone acetate (−6.314 kcal/mol) with a relatively strong binding affinity, respectively. Importantly, CNNE2 protein level could be upregulated with EPT and attenuated by estrogen receptor antagonist, acolbifene and had interactions with cancer driver genes (AKT1 and KRAS) and high mutation frequency gene (TP53 and PTEN) in breast cancer patients. In conclusion, the current study showed that CCNE2, CDCA5, RAD51, TCF19, KNTC1, MCM10, and NEIL3 might contribute to EPT-related tumorigenesis in breast cancer, with CCNE2 might be a sensitive risk indicator of breast cancer risk in women using MHT.
Collapse
Affiliation(s)
- Yu Deng
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing 100034, China
| | - He Huang
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing 100034, China
| | - Jiangcheng Shi
- School of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Hongyan Jin
- Department of Obstetrics and Gynecology, Peking University First Hospital, No. 8 Xishiku Street, Beijing 100034, China
- Correspondence:
| |
Collapse
|
17
|
Effect of CDCA5 on Proliferation and Metastasis of Triple Negative Breast Cancer Cells under shRNA Interference Technology. JOURNAL OF ONCOLOGY 2022; 2022:9038230. [PMID: 35726220 PMCID: PMC9206565 DOI: 10.1155/2022/9038230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/24/2022] [Accepted: 03/05/2022] [Indexed: 11/25/2022]
Abstract
Objective It was to explore the effect of cell division cycle associated 5 (CDCA5) under shRNA interference on proliferation and metastasis of triple negative breast cancer (TNBC) cells. Methods MDA-ME-231 and BT549 cells were selected as the research objects. According to the different interference methods and CDCA5 interference sequences, they were divided into the interference group 1MDA-ME-231, the interference group 2MDA-ME-231, the interference group 1BT549, the interference group 2BT549 (using shRNA technology), the control group MDA-ME-231, and the control group BT549 (breast cancer cells under normal culture conditions). MCF10A cells were routinely cultured as the negative control group to analyze the effect of CDCA5 expression on the proliferation and migration of cancer cells. Results The expression of CDCA5 protein in MDA-ME-231 and BT549 cells in control group was significantly higher than that in negative control group (P < 0.05). Compared with the control group, the inhibition rates of CDCA5 expression in 1MDA-ME-231, 2MDA-ME-231, 1BT549, and 2BT549 cells in the interference group were 39.01%, 42.98%, 49.57%, and 60.98%, respectively (P < 0.05). From 12 h, the proliferation level of TNBC cells at different culture time was lower than that of the control group (P < 0.05). Compared with the number of staining cells in the control group, the positive staining cells in 1MDA-ME-231 (61.42%), 2MDA-ME-231 (72.06%), 1BT549 (52.53%), and 2BT549 (59.65%) in the interference group were significantly decreased (P < 0.05). Conclusion The results show that the expression of CDCA5 in TNBC is increased, which plays an important role in the proliferation and migration of cancer cells. shRNA interference technology can knock down the expression of CDCA5 and inhibit its “promoting cancer” effect.
Collapse
|
18
|
Huang X, Huang Y, Lv Z, Wang T, Feng H, Wang H, Du S, Wu S, Shen D, Wang C, Li H, Wang B, Ma X, Zhang X. Loss of cell division cycle‑associated 5 promotes cell apoptosis by activating DNA damage response in clear cell renal cell carcinoma. Int J Oncol 2022; 61:87. [PMID: 35642672 PMCID: PMC9183765 DOI: 10.3892/ijo.2022.5377] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/18/2022] [Indexed: 12/24/2022] Open
Abstract
Cell division cycle-associated 5 (CDCA5) protein, which is involved in cohesion, contributes to cell cycle regulation and chromosome segregation by maintaining genomic stability. Accumulating evidence indicates that CDCA5 expression is upregulated in a number of types of cancer associated with a poor prognosis. However, the biological function of CDCA5 in clear cell renal cell carcinoma (ccRCC) remains largely unknown. In the present study, The Cancer Genome Atlas data mining revealed that CDCA5 was more highly expressed in ccRCC than in adjacent normal tissues. Importantly, such a high expression was associated with a higher risk of distant metastasis and poorer clinical outcomes. Moreover, the clinical and prognostic value of CDCA5 expression was further investigated using immunohistochemistry on tissue microarrays containing paired tumor tissues and adjacent normal tissues from 137 patients with ccRCC. Functional analyses revealed that CDCA5 knockdown significantly inhibited the proliferation and migration of ccRCC cells, and suppressed the growth of xenografts in nude mice. Mechanistically, CDCA5 knockdown induced severe DNA damage with the persistent accumulation of γ-H2A histone family member X foci, resulting in G2/M cell cycle arrest and finally, in chromosomal instability and apoptosis. CDCA5 knockdown significantly decreased the phosphorylation levels of Stat3 and NF-κB, suggesting that CDCA5 plays a role in regulating the inflammatory response. Collectively, the findings of the present study indicate that ccRCC cells require CDCA5 for malignant progression, and that CDCA5 inhibition may enhance the outcomes of patients with high-risk ccRCC.
Collapse
Affiliation(s)
- Xing Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Yan Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Zheng Lv
- Department of Urology, The Third Affiliated Central Hospital of Nankai University, Tianjin 300071, P.R. China
| | - Tao Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Huayi Feng
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Hanfeng Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Songliang Du
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Shengpan Wu
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Donglai Shen
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Chenfeng Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Hongzhao Li
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Baojun Wang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Xin Ma
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Xu Zhang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital/Medical School of Chinese PLA/State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
19
|
Li J, Liu L, Chen Y, Wu M, Lin X, Shen Z, Cheng Y, Chen X, Weygant N, Wu X, Wei L, Sferra TJ, Han Y, Chen X, Shen A, Shen A, Peng J. Ribosome assembly factor PNO1 is associated with progression and promotes tumorigenesis in triple‑negative breast cancer. Oncol Rep 2022; 47:108. [PMID: 35445733 PMCID: PMC9073417 DOI: 10.3892/or.2022.8319] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/14/2022] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to investigate the expression of ribosome assembly factor partner of NOB1 homolog (PNO1) and its association with the progression of breast cancer (BC) in patients, as well as its biological function and underlying mechanism of action in BC cells. Bioinformatics and immunohistochemical analyses revealed that PNO1 expression was significantly increased in BC tissues and its high mRNA expression was associated with shorter overall survival (OS) and relapse-free survival (RFS) of patients with BC, as well as multiple clinical characteristics (including advanced stage of NPI and SBR, etc.) of patients with BC. Biological functional studies revealed that transduction of lentivirus encoding sh-PNO1 significantly downregulated PNO1 expression, reduced cell confluency and the number of BC cells in vitro and inhibited tumor growth in vivo. Moreover, PNO1 knockdown decreased the cell viability and arrested cell cycle progression at the G2/M phase, as well as downregulated cyclin B1 (CCNB1) and cyclin-dependent kinase 1 (CDK1) protein expression in BC cells. Correlation analysis demonstrated that PNO1 expression was positively correlated with both CDK1 and CCNB1 expression in BC samples. Collectively, PNO1 was upregulated in BC and associated with BC patient survival, and PNO1 knockdown suppressed tumor growth in vitro and in vivo. In addition, positive regulation of CCNB1 and CDK1 may be one of the underlying mechanisms.
Collapse
Affiliation(s)
- Jie Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiaoying Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Zhiqing Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiaoping Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiangyan Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Thomas J Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, OH 44106, USA
| | - Yuying Han
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xi Chen
- Department of Oncology, No. 900 Hospital of The Joint Logistic Support Force, Fuzhou, Fujian 350025, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
20
|
Shen W, Tong D, Chen J, Li H, Hu Z, Xu S, He S, Ge Z, Zhang J, Mao Q, Chen H, Xu G. Silencing oncogene cell division cycle associated 5 induces apoptosis and G1 phase arrest of non‐small cell lung cancer cells via p53‐p21 signaling pathway. J Clin Lab Anal 2022; 36:e24396. [PMID: 35373420 PMCID: PMC9102649 DOI: 10.1002/jcla.24396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Wei Shen
- Department of Pulmonary and Critical Care Medicine The Third People’s Hospital of Cixi Ningbo Zhejiang China
| | - Dimin Tong
- Department of Pulmonary and Critical Care Medicine The Third People’s Hospital of Cixi Ningbo Zhejiang China
| | - Jie Chen
- Department of Pulmonary and Critical Care Medicine The Third People’s Hospital of Cixi Ningbo Zhejiang China
| | - Hongxiang Li
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Zeyang Hu
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Shuguang Xu
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Sufang He
- Department of Pulmonary and Critical Care Medicine Guangdong Provincial People's Hospital Ganzhou Hospital Ganzhou Jiangxi China
| | - Zhen Ge
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Jianan Zhang
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Qiqi Mao
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Hang Chen
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| | - Guodong Xu
- Department of Cardiothoracic Surgery The Affiliated Lihuili Hospital Ningbo University Ningbo Zhejiang China
| |
Collapse
|
21
|
Yu Z, Wu J, Zhang L, Liu SY. Potential molecular target screening and bioinformatics analysis of cholangiocarcinoma based on GEO database. Shijie Huaren Xiaohua Zazhi 2022; 30:128-135. [DOI: 10.11569/wcjd.v30.i3.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma is a highly malignant tumor with a poor prognosis. Targeted therapy is important for the treatment of cholangiocarcinoma, and it is therefore of great clinical importance to identify novel molecular targets for targeted therapy of this malignancy.
AIM To identify potential molecular targets for the treatment of cholangiocarcinoma and identify the key genes involved in cholangiocarcinoma by bioinformatics analysis.
METHODS We downloaded two sets of cholangiocarcinoma expression profile data from GEO database. GEO2R online analysis tool was used to screen differentially expressed genes in cholangiocarcinoma tumor tissues and normal tissues, and we performed GO enrichment analysis, KEGG pathway analysis, and protein interaction network for differentially expressed genes. We used Cytoscape software to calculate key genes. The GEPIA database was used to verify the expression of hub genes in cholangiocarcinoma tissues.
RESULTS A total of 158 differentially expressed genes were identified. GO enrichment analysis showed that these differential genes were mainly involved in the cellular response to zinc ion, negative regulation of growth, cell adhesion, metabolic process, and protein homotetramerization. They were enriched in exosomes, extracellular spaces, elastic fibers, and organelle membranes. The main molecular functions are related to heparin binding, cysteine-type endopeptidase inhibitor activity, protein homodimerization activity, receptor binding, and pyridoxal phosphate binding. KEGG pathway analysis showed that differential genes are mainly involved in processes such as mineral absorption, carbon and propanoate metabolism, PPAR signaling pathway, and fatty acid degradation. A protein interaction network diagram was constructed based on the String database, and the CytoHubba plug-in of the Cytoscape software was used to calculate the key genes. The key genes were all up-regulated ones. GEPIA analysis verified that the expression of key genes in cholangiocarcinoma tissues was significantly higher than that in normal tissues.
CONCLUSION In this study, eight key genes related to cholangiocarcinoma were identified, including NUSAP1, TOP2A, RAD51AP1, MCM4, KIAA0101, CDCA5, TYMS, and ZWINT. These genes provide new ideas for in-depth study of the targeted therapy of cholangiocarcinoma, and are expected to become new molecular therapeutic targets.
Collapse
Affiliation(s)
- Zhen Yu
- Department of Laboratory Medicine, The Third Central Hospital of Tianjin; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China
| | - Jing Wu
- Department of Laboratory Medicine, The Third Central Hospital of Tianjin; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China
| | - Lei Zhang
- Department of Laboratory Medicine, The Third Central Hospital of Tianjin; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China
| | - Shu-Ye Liu
- Department of Laboratory Medicine, The Third Central Hospital of Tianjin; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin 300170, China
| |
Collapse
|
22
|
Luo Z, Hu Q, Tang Y, Leng Y, Tian T, Tian S, Huang C, Liu A, Deng X, Shen L. Construction and investigation of β3GNT2-associated regulatory network in esophageal carcinoma. Cell Mol Biol Lett 2022; 27:8. [PMID: 35073841 PMCID: PMC8903709 DOI: 10.1186/s11658-022-00306-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/04/2022] [Indexed: 12/24/2022] Open
Abstract
Background Glycosyltransferases play a crucial role in various cancers. β1, 3-N-acetylglucosaminyltransferase 2, a polylactosamine synthase, is an important member of the glycosyltransferase family. However, the biological function and regulatory mechanism of β3GNT2 in esophageal carcinoma (ESCA) is still poorly understood. Methods The Cancer Genome Atlas and Genotype-Tissue Expression databases were used for gene expression and prognosis analysis. Quantitative real-time PCR, Western blot, and immunohistochemistry were performed to detect the expression of β3GNT2 in ESCA cell lines and tissues. In vitro assays and xenograft tumor models were utilized to evaluate the impact of β3GNT2 on ESCA progression. The downstream effectors and upstream regulators of β3GNT2 were predicted by online software and verified by functional experiments. Results We found that β3GNT2 was highly expressed in ESCA tissues and positively correlated with poor prognosis in ESCA patients. β3GNT2 expression was closely associated with the tumor size, TNM stage, and overall survival of ESCA patients. Functionally, β3GNT2 promoted ESCA cell growth, migration, and invasion in vitro, as well as tumorigenesis in vivo. Mechanistically, β3GNT2 knockdown decreased the expression of the polylactosamine on EGFR. Knockdown of β3GNT2 also inhibited the JAK/STAT signaling pathway. Meanwhile, the JAK/STAT inhibitor could partly reverse the biological effects caused by β3GNT2 overexpression. Moreover, β3GNT2 expression was positively regulated by CREB1 and negatively regulated by miR-133b. Both CREB1 and miR-133b was involved in the β3GNT2-mediated ESCA progression. Conclusions Our study, for the first time, reveals the importance of β3GNT2 in ESCA progression and offers a potential therapeutic target for ESCA. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00306-y.
Collapse
|
23
|
Chen R, Ma S, Qiao H, Su F, Wang L, Guan Q. Identification of target genes and prognostic evaluation for colorectal cancer using integrated bioinformatics analysis. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2026825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Rui Chen
- Department of the First Clinical Medical College, Lanzhou University, Lanzhou, People’s Republic of China
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| | - Shoucheng Ma
- Department of the First Clinical Medical College, Lanzhou University, Lanzhou, People’s Republic of China
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| | - Hui Qiao
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| | - Fei Su
- Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| | - Lina Wang
- Department of the First Clinical Medical College, Lanzhou University, Lanzhou, People’s Republic of China
- Department of Radiotherapy, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| | - QuanLin Guan
- Department of the First Clinical Medical College, Lanzhou University, Lanzhou, People’s Republic of China
- Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| |
Collapse
|
24
|
Pan J, Huang G, Yin Z, Cai X, Gong E, Li Y, Xu C, Ye Z, Cao Z, Cheng W. Circular RNA FLNA acts as a sponge of miR-486-3p in promoting lung cancer progression via regulating XRCC1 and CYP1A1. Cancer Gene Ther 2022; 29:101-121. [PMID: 33500536 PMCID: PMC8761575 DOI: 10.1038/s41417-021-00293-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/18/2020] [Accepted: 01/05/2021] [Indexed: 01/29/2023]
Abstract
Significantly high-expressed circFLNA has been found in various cancer cell lines, but not in lung cancer. Therefore, this study aimed to explore the role of circFLNA in the progression of lung cancer. The target gene of circFLNA was determined by bioinformatics and luciferase reporter assay. Viability, proliferation, migration, and invasion of the transfected cells were detected by CCK-8, colony formation, wound-healing, and transwell assays, respectively. A mouse subcutaneous xenotransplanted tumor model was established, and the expressions of circFLNA, miR-486-3p, XRCC1, CYP1A1, and related genes in the cancer cells and tissues were detected by RT-qPCR, Western blot, or immunohistochemistry. The current study found that miR-486-3p was low-expressed in lung cancer. MiR-486-3p, which has been found to target XRCC1 and CYP1A1, was regulated by circFLNA. CircFLNA was located in the cytoplasm and had a high expression in lung cancer cells. Cancer cell viability, proliferation, migration, and invasion were promoted by overexpressed circFLNA, XRCC1, and CYP1A1 but inhibited by miR-486-3p mimic and circFLNA knockdown. The weight of the xenotransplanted tumor was increased by circFLNA overexpression yet reduced by miR-486-3p mimic. Furthermore, miR-486-3p mimic reversed the effect of circFLNA overexpression on promoting lung cancer cells and tumors and regulating the expressions of miR-486-3p, XRCC1, CYP1A1, and metastasis/apoptosis/proliferation-related factors. However, overexpressed XRCC1 and CYP1A1 reversed the inhibitory effect of miR-486-3p mimic on cancer cells and tumors. In conclusion, circFLNA acted as a sponge of miR-486-3p to promote the proliferation, migration, and invasion of lung cancer cells in vitro and in vivo by regulating XRCC1 and CYP1A1.
Collapse
Affiliation(s)
- Jiongwei Pan
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Gang Huang
- Department of Chinese Medicine, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Zhangyong Yin
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Xiaoping Cai
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Enhui Gong
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Yuling Li
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Cunlai Xu
- Department of Respiratory, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Zaiting Ye
- Department of Radiology, the Sixth Affiliated Hospital of Wenzhou Medical University/Lishui People's Hospitlal, Lishui, Zhejiang, 323000, China
| | - Zhuo Cao
- The Sixth Affiliated Hospital of Wenzhou Medical University; Longquan Branch, Lishui People's Hospitlal, Lishui, China.
| | - Wei Cheng
- Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, Jiangsu Province Key Laboratory of Anesthesiology and Center for Pain Research and Treatment, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
25
|
Luo Z, Wang J, Zhu Y, Sun X, He C, Cai M, Ma J, Wang Y, Han S. SPOP promotes CDCA5 degradation to regulate prostate cancer progression via the AKT pathway. Neoplasia 2021; 23:1037-1047. [PMID: 34509929 PMCID: PMC8435818 DOI: 10.1016/j.neo.2021.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 12/02/2022]
Abstract
The E3 ubiquitin ligase adaptor Speckle-type POZ protein (SPOP) plays an important tumour suppressor role in prostate cancers (PCa), with mutation rate up to 15%. However, how SPOP mutations regulate prostate tumorigenesis remains elusive. Here, we report the identification of cell division cycle associated 5 (CDCA5) as a SPOP substrate. We found that SPOP interacts with CDCA5 and promotes its polyubiquitin degradation in a degron-dependent manner. This effect was greatly impaired by introducing PCa associated SPOP mutations. Importantly, we found that CDCA5 was essential for PCa cells to survive and proliferate. CDCA5 depletion in PCa cells led to cessation of proliferation, G2M arrest, severe sister chromatid aggregation disturbance, and apoptosis. we also found that CDCA5 knockdown decreased the protein expression of p-GSK3β, increased the activity of caspase-3, caspase-9, and the Bax/Bcl-2 ratio. Besides, we confirmed that CDCA5 interrupted cancer cell behavior via the AKT pathway. In contrast, silencing SPOP or overexpressing CDCA5 increased cell proliferation. Consistently, depleting SPOP along with CDCA5, or overexpressing CDCA5 along with SPOP also caused the growth of cells repressed. Consistent with the functional role of CDCA5, the mRNA and protein levels of CDCA5 were significantly increased in PCa, compared to normal tissues, and its high expression was associated with more severe lymph node metastasis, higher Gleason score, and poorer prognosis. Together, our data showed that SPOP plays a crucial role in inhibiting tumorigenesis and partly achieved this by promoting the degradation of oncoprotein CDCA5.
Collapse
Affiliation(s)
- Zhenzhen Luo
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Zhu
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiao Sun
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chenchen He
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjiao Cai
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jinlu Ma
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| | - Suxia Han
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
26
|
Laliotis GI, Chavdoula E, Paraskevopoulou MD, Kaba A, La Ferlita A, Singh S, Anastas V, Nair KA, Orlacchio A, Taraslia V, Vlachos I, Capece M, Hatzigeorgiou A, Palmieri D, Tsatsanis C, Alaimo S, Sehgal L, Carbone DP, Coppola V, Tsichlis PN. AKT3-mediated IWS1 phosphorylation promotes the proliferation of EGFR-mutant lung adenocarcinomas through cell cycle-regulated U2AF2 RNA splicing. Nat Commun 2021; 12:4624. [PMID: 34330897 PMCID: PMC8324843 DOI: 10.1038/s41467-021-24795-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
AKT-phosphorylated IWS1 regulates alternative RNA splicing via a pathway that is active in lung cancer. RNA-seq studies in lung adenocarcinoma cells lacking phosphorylated IWS1, identified a exon 2-deficient U2AF2 splice variant. Here, we show that exon 2 inclusion in the U2AF2 mRNA is a cell cycle-dependent process that is regulated by LEDGF/SRSF1 splicing complexes, whose assembly is controlled by the IWS1 phosphorylation-dependent deposition of histone H3K36me3 marks in the body of target genes. The exon 2-deficient U2AF2 mRNA encodes a Serine-Arginine-Rich (RS) domain-deficient U2AF65, which is defective in CDCA5 pre-mRNA processing. This results in downregulation of the CDCA5-encoded protein Sororin, a phosphorylation target and regulator of ERK, G2/M arrest and impaired cell proliferation and tumor growth. Analysis of human lung adenocarcinomas, confirmed activation of the pathway in EGFR-mutant tumors and showed that pathway activity correlates with tumor stage, histologic grade, metastasis, relapse after treatment, and poor prognosis.
Collapse
Affiliation(s)
- Georgios I Laliotis
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA.
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA.
- School of Medicine, University of Crete, Heraklion, Crete, Greece.
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Evangelia Chavdoula
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | | | - Abdul Kaba
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Alessandro La Ferlita
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Satishkumar Singh
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
- Department of Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Vollter Anastas
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
- Tufts Graduate School of Biomedical Sciences, Program in Genetics, Boston, MA, USA
| | - Keith A Nair
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Arturo Orlacchio
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Vasiliki Taraslia
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis Vlachos
- DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
- Department Of Pathology, Beth Israel-Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marina Capece
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | | | - Dario Palmieri
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Christos Tsatsanis
- School of Medicine, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
| | - Salvatore Alaimo
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Lalit Sehgal
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
- Department of Medicine, Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - David P Carbone
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Philip N Tsichlis
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA.
- The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA.
- Tufts Graduate School of Biomedical Sciences, Program in Genetics, Boston, MA, USA.
| |
Collapse
|
27
|
Li Z, Liu Z, Li C, Liu Q, Tan B, Liu Y, Zhang Y, Li Y. CDCA1/2/3/5/7/8 as novel prognostic biomarkers and CDCA4/6 as potential targets for gastric cancer. Transl Cancer Res 2021; 10:3404-3417. [PMID: 35116645 PMCID: PMC8798863 DOI: 10.21037/tcr-20-1050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/10/2021] [Indexed: 01/09/2023]
Abstract
Background Increasing evidence had suggested that cell division cycle-associated (CDCA) family proteins play prominent roles in multiple types of cancer. However, the expression pattern and prognostic value of CDCAs in gastric cancer were still poorly understood. Methods In this study, bioinformatics was used for the first time to comprehensively discuss the expression changes of the CDCA protein family in gastric cancer. We studied the transcription and survival data of CDCAs in patients with gastric cancer in Oncomine, GEPIA, DAVID, cBioportal, and other databases. Results We identified that the CDCA 1/2/3/4/5/6/7/8 were overexpressed gastric cancer than in normal tissues. There was no significant difference in CDCAs expression among different gastric cancer stages. High expression of CDCA4/6 in patients with gastric cancer was closely related to low overall survival (OS), first progression survival (FPS), and post-progression survival (PPS). In contrast, high CDCA1/2/3/5/7/8 expression predicted a better prognosis. The genetic mutation rate of CDCA2 and CDCA4 was 4%, ranking first. The main biological process of CDCAs protein family enrichment was cell division, the main cell component involved was centromeres of chromosomes, and the main molecular function involved was protein binding. Conclusions The study suggested that CDCA1/2/3/5/7/8 were expected to be new prognostic markers for gastric cancer, and CDCA4/6 might be potential targets for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Zhaoxing Li
- Department of General Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital and Institute, Beijing, China
| | - Chuang Li
- The Second Hospital of Shijiazhuang, Shijiazhuang, China
| | - Qingwei Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bibo Tan
- Department of General Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yu Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Yong Li
- Department of General Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
28
|
Markiewicz LH, Ogrodowczyk AM, Wiczkowski W, Wróblewska B. Phytate and Butyrate Differently Influence the Proliferation, Apoptosis and Survival Pathways in Human Cancer and Healthy Colonocytes. Nutrients 2021; 13:1887. [PMID: 34072741 PMCID: PMC8230256 DOI: 10.3390/nu13061887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/19/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
The colonic epithelium is never exposed to a single factor, therefore studies on the effect of combinations of factors naturally and persistently present in the intestines are of special importance for understanding the phenomena occurring at this place. The aim of the study was to investigate the combined effect of 1 mM phytate and 1 mM butyrate (PA1B1) on cell lines derived from cancer (HCT116 and HT-29) and healthy (NCM460D) human colonic epithelium. Colorimetric and flow cytometry methods were used to determine the proliferation rate, cell cycle, and apoptosis. Selected markers of proliferation, inflammatory, and survival pathways were investigated at the mRNA and/or protein level. The combination of phytate and butyrate disturbed the cell cycle and triggered apoptosis and/or death in both studied cancer colonocytes to a higher extent compared to healthy colonocytes. Moreover, in healthy colonocytes, phytate activated the survival pathway without stimulation of inflammatory response. This may indicate that the response of healthy colonocytes to phytate protects colonic epithelium from the loss of integrity and tightness that would occur if inflammation developed. Based on the obtained results we postulate that studies on both cancer and/or healthy colonocytes should be carried out in the presence of butyrate as the permanent component of colonic contents. This should be of special importance when anti-proliferative/pro-apoptotic activity or inflammatory status of colonocytes is to be investigated.
Collapse
Affiliation(s)
- Lidia Hanna Markiewicz
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (A.M.O.); (B.W.)
| | - Anna Maria Ogrodowczyk
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (A.M.O.); (B.W.)
| | - Wiesław Wiczkowski
- Department of Chemistry and Biodynamics of Food, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland;
| | - Barbara Wróblewska
- Department of Immunology and Food Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland; (A.M.O.); (B.W.)
| |
Collapse
|
29
|
Zhang YF, Wang YX, Zhang N, Lin ZH, Wang LR, Feng Y, Pan Q, Wang L. Prognostic alternative splicing regulatory network of RBM25 in hepatocellular carcinoma. Bioengineered 2021; 12:1202-1211. [PMID: 33830865 PMCID: PMC8806338 DOI: 10.1080/21655979.2021.1908812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
RNA-binding motif protein 25 (RBM25) is a poorly characterized RNA-binding protein that is involved in several biological processes and regulates the proliferation and metastasis of tumor cells. The regulatory role of RBM25 in hepatocellular carcinoma (HCC) is unknown. Here, RBM25 expression and outcomes in HCC patients were evaluated using The Cancer Genome Atlas database. RBM25 was overexpressed in HCC patients compared with the healthy group. The high expression of RBM25 in tumor tissues was significantly related to poor overall survival (P<0.001). Overexpression of RBM25 significantly contributed to poorer survival in male patients and N0 stage patients (P<0.001). Spearman analysis and weighted gene co-expression network analysis identified 694 RBM25-related genes. Protein-protein interaction network analysis revealed the Cluster with the highest score, which positively correlated with RBM25. CDCA5 and INCENP were identified as the core functional genes related to RBM25. The overexpression of CDCA5 and INCENP in HCC patients was examined using the Human Protein Atlas database. The findings collectively indicated that RBM25 may interact with CDCA5 and INCENP to regulate HCC. Our detailed characterization of RBM25 protein interactions and related core functional genes provides a basis for further studies aimed at identifying molecular regulatory pathways or splicing events.
Collapse
Affiliation(s)
- Yong-Fa Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Xiu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning- Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen-Hai Lin
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Long-Rong Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun Feng
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Pan
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
LINC01515 promotes nasopharyngeal carcinoma progression by serving as a sponge for miR-325 to up-regulate CDCA5. J Mol Histol 2021; 52:577-587. [PMID: 33770322 DOI: 10.1007/s10735-021-09969-x] [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] [Received: 11/17/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022]
Abstract
Long non-coding RNAs (LncRNAs) have gained widespread interest and attention as vital regulators in cancer occurrence and development. Nonetheless, the functions and mechanisms of lncRNAs involved in nasopharyngeal carcinoma (NPC) are largely unknown. By analysing the data from GSE61218, we identified a novel lncRNA LINC01515 which is altered in NPC. A series of experiments were performed to examine the exact roles of LINC01515 as well as the molecular mechanisms by which LINC01515 acted in NPC. LINC01515 expression was increased in NPC and that high LINC01515 expression was associated with a worse prognosis. Functionally, depletion of LINC01515 resulted in an inhibition of cell proliferation, migration and invasion, while apoptosis was promoted. Mechanistically, LINC01515 facilitated cell division cycle associated 5 (CDCA5) expression via serving as a sponge for miR-325. And more notably, miR-325 suppressed NPC progression in vitro by targeting CDCA5. Furthermore, the anti-tumor property induced by LINC01515 knockdown was partially reversed due to the overexpression of CDCA5. Taken together, LINC01515 exerted the carcinogenic effect in NPC through regulating miR-325/CDCA5 pathway. Our findings shed light on the possibility of exploiting LINC01515 as a prognostic biomarker or therapeutic target in NPC.
Collapse
|
31
|
Wang Z, Xu J, Zhang S, Chang L. Expression of cell divisioncycle-associated genes and their prognostic significance in hepatocellular carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:151-169. [PMID: 33564348 PMCID: PMC7868782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
The cell division cycle-associated (CDCA) protein family plays an essential role in tumor progression by cell division. However, the function of each CDCA family member in hepatocellular carcinoma (HCC) is not well known. This study is to find the roles of CDCAs in the prognosis of HCC patients by using ONCOMINE, UALCAN, Human Protein Atlas, Kaplan-Meier Plotter, and cBioPortal databases. Overexpression of CDCA mRNA and protein were found to be significantly associated with individual cancer stages and tumor grades in HCC patients. Higher mRNA expressions of 6 CDCA family members were found to be significantly associated with shorter overall survival (OS) in HCC patients. Multivariate analysis showed that overexpressions of CDCA mRNA were independent prognostic factors for shorter OS in HCC patients. Moreover, a high mutation rate of CDCAs (27%) was also detected in HCC patients, and genetic alteration in CDCAs was associated with shorter overall survival (OS) and disease-free survival (DFS) in HCC patients. Finally, a functional analysis showed that CDCAs were mainly enriched in the cell cycle (hsa04110) and oocyte meiosis. Overall, these results indicated that CDCA2/3/4/5/8 could be prognostic biomarkers of survival in HCC patients.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Gastroenterology, Shijiazhuang People’s HospitalShijiazhuang, Hebei, P. R. China
| | - Jianduo Xu
- Department of General Surgery, Shijiazhuang People’s HospitalShijiazhuang, Hebei, P. R. China
| | - Shumei Zhang
- Department of Gastroenterology, Shijiazhuang People’s HospitalShijiazhuang, Hebei, P. R. China
| | - Lili Chang
- Department of Gastroenterology, Shijiazhuang People’s HospitalShijiazhuang, Hebei, P. R. China
| |
Collapse
|
32
|
Transcriptome Profiling of Human Follicle Dermal Papilla Cells in response to Porphyra-334 Treatment by RNA-Seq. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6637513. [PMID: 33519944 PMCID: PMC7817261 DOI: 10.1155/2021/6637513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/18/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
Porphyra-334 is a kind of mycosporine-like amino acid absorbing ultraviolet-A. Here, we characterized porphyra-334 as a potential antiaging agent. An in vitro assay revealed that porphyra-334 dramatically promoted collagen synthesis in fibroblast cells. The effect of porphyra-334 on cell proliferation was dependent on the cell type, and the increase of cell viability by porphyra-334 was the highest in keratinocyte cells among the three tested cell types. An in vivo clinical test with 22 participants demonstrated the possible role of porphyra-334 in the improvement of periorbital wrinkles. RNA-sequencing using human follicle dermal papilla (HFDP) cells upon porphyra-334 treatment identified the upregulation of metallothionein- (MT-) associated genes, confirming the antioxidant role of porphyra-334 with MT. Moreover, the expression of genes involved in nuclear chromosome segregation and the encoding of components of kinetochores was upregulated by porphyra-334 treatment. Furthermore, we found that several genes associated with the hair follicle cycle, the hair follicle structure, the epidermal structure, and stem cells were upregulated by porphyra-334 treatment, suggesting the potential role of porphyra-334 in hair follicle growth and maintenance. In summary, we provided several new pieces of evidence of porphyra-334 as a potential antiaging cosmetic agent and elucidated the expression network in HFDP cells upon porphyra-334.
Collapse
|
33
|
Ji J, Shen T, Li Y, Liu Y, Shang Z, Niu Y. CDCA5 promotes the progression of prostate cancer by affecting the ERK signalling pathway. Oncol Rep 2021; 45:921-932. [PMID: 33650660 PMCID: PMC7859918 DOI: 10.3892/or.2021.7920] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Cell division cycle-associated 5 (CDCA5) can regulate cell cycle-related proteins to promote the proliferation of cancer cells. The purpose of the present study was to investigate the expression level of CDCA5 in prostate cancer (PCa) and its effect on PCa progression. The signalling pathway by which CDCA5 functions through was also attempted to elucidate. Clinical specimens of PCa patients were collected from the Second Hospital of Tianjin Medical University. The expression level of CDCA5 in cancer tissues and paracancerous tissues from PCa patients was detected by RT-qPCR analysis and IHC. The relationship between the expression level of CDCA5 and the survival rate of PCa patients was analysed using TCGA database. Two stable cell lines (C4-2 and PC-3) with CDCA5 knockdown were established, and the effects of CDCA5 on PCa cell proliferation were detected by MTT and colony formation assays. Flow cytometry was performed to detect the effect of CDCA5 on the PCa cell division cycle, and western blot analysis was used to determine changes in ERK phosphorylation levels after CDCA5 knockdown. The effect of CDCA5 expression on prostate tumour growth was assessed using a mouse xenograft model. The results revealed that the mRNA and protein expression levels of CDCA5 were significantly higher in PCa tissues than in paracancerous tissues. High CDCA5 expression was associated with the prognosis of patients with PCa. CDCA5 expression knockdown significantly reduced the number of PCa cells in mitoses and inhibited their proliferation in vitro and in vivo. When CDCA5 was knocked down, the phosphorylation level of ERK was also reduced. Collectively, CDCA5 was upregulated and affected the prognosis of patients with PCa. Decreased CDCA5 expression inhibited PCa cell proliferation by inhibiting the ERK signalling pathway. Thus, CDCA5 may be a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Junpeng Ji
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Tianyu Shen
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Yang Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Yixi Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Zhiqun Shang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| | - Yuanjie Niu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Hexi, Tianjin 300211, P.R. China
| |
Collapse
|
34
|
Wu D, Zhong P, Wang Y, Zhang Q, Li J, Liu Z, Ji A, Li Y. Hydrogen Sulfide Attenuates High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease by Inhibiting Apoptosis and Promoting Autophagy via Reactive Oxygen Species/Phosphatidylinositol 3-Kinase/AKT/Mammalian Target of Rapamycin Signaling Pathway. Front Pharmacol 2020; 11:585860. [PMID: 33390956 PMCID: PMC7774297 DOI: 10.3389/fphar.2020.585860] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease worldwide. Hydrogen sulfide (H2S) is involved in a wide range of physiological and pathological processes. Nevertheless, the mechanism of action of H2S in NAFLD development has not been fully clarified. Here, the reduced level of H2S was observed in liver cells treated with oleic acid (OA). Administration of H2S increased the proliferation of OA-treated cells. The results showed that H2S decreased apoptosis and promoted autophagy through reactive oxygen species (ROS)-mediated phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) cascade in OA-treated cells. In addition, administration of H2S relieved high-fat diet (HFD)-induced NAFLD via inhibition of apoptosis and promotion of autophagy. These findings suggest that H2S could ameliorate HFD-induced NAFLD by regulating apoptosis and autophagy through ROS/PI3K/AKT/mTOR signaling pathway. Novel H2S-releasing donors may have therapeutic potential for the treatment of NAFLD.
Collapse
Affiliation(s)
- Dongdong Wu
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China.,School of Stomatology, Henan University, Kaifeng, China
| | - Peiyu Zhong
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Yizhen Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Qianqian Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Jianmei Li
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Zhengguo Liu
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Ailing Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Yanzhang Li
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| |
Collapse
|
35
|
Lin K, Zhu X, Luo C, Bu F, Zhu J, Zhu Z. Data mining combined with experiments to validate CEP55 as a prognostic biomarker in colorectal cancer. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:167-182. [PMID: 33190424 PMCID: PMC7860595 DOI: 10.1002/iid3.375] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a common tumor with high morbidity and mortality. Current specific diagnosis regarding CRC remains complicated and costly, and specific diagnostic biomarkers are lacking. METHODS To find potential diagnostic and prognostic biomarkers for CRC, we screened and analyzed many CRC sequencing data by The Cancer Genome Atlas Program and Gene Expression Omnibus, and validated that CEP55 may be a potential diagnostic biomarker for CRC by molecular cytological experiments and immunohistochemistry, among others. RESULTS We found that CEP55 is upregulated in CRC tissues and tumor cells and can promote CRC proliferation and metastasis by activating the p53/p21 axis and that CEP55 mutations in tumor patients result in worse overall survival and disease-free survival time. Besides, we also found that genes, such as CDK1, CCNB1, NEK2, KIF14, CDCA5, and RFC3 were upregulated in tumors, and their mutations would affect the prognosis of CRC patients, but these results await for more experimental evidence. CONCLUSION Our study validates CEP55 as a potential diagnostic and prognostic biomarker for CRC, and we also provide multiple genes and potential molecular mechanisms that may serve as diagnostic and prognostic markers for CRC.
Collapse
Affiliation(s)
- Kang Lin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chen Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fanqin Bu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jinfeng Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhengming Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
36
|
Yi M, Li T, Qin S, Yu S, Chu Q, Li A, Wu K. Identifying Tumorigenesis and Prognosis-Related Genes of Lung Adenocarcinoma: Based on Weighted Gene Coexpression Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4169691. [PMID: 32149105 PMCID: PMC7035528 DOI: 10.1155/2020/4169691] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/18/2020] [Indexed: 02/07/2023]
Abstract
Lung adenocarcinoma is the most frequently diagnosed subtype of nonsmall cell lung cancer. The molecular mechanisms of the initiation and progression of lung adenocarcinoma remain to be further determined. This study aimed to screen genes related to the progression of lung adenocarcinoma. By weighted gene coexpression network analysis (WGCNA), we constructed a free-scale gene coexpression network to evaluate the correlations between multiple gene sets and patients' clinical traits, then further identify predictive biomarkers. GSE11969 was obtained from the Gene Expression Omnibus (GEO) database which contained the gene expression data of 90 lung adenocarcinoma patients. Data of the Cancer Genome Atlas (TCGA) were employed as the validation cohort. After the average linkage hierarchical clustering, a total of 9 modules were generated. In the clinical significant module (R = 0.44, P < 0.0001), we identified 29 network hub genes. Subsequent verification in the TCGA database showed that 11 hub genes (ANLN, CDCA5, FLJ21924, LMNB1, MAD2L1, RACGAP1, RFC4, SNRPD1, TOP2A, TTK, and ZWINT) were significantly associated with poor survival data of lung adenocarcinomas. Besides, the results of receiver operating characteristic curves indicated that the mRNA levels of this group of genes exhibited high specificity and sensitivity to distinguish malignant lesions from nonmalignant tissues. Apart from mRNA levels, we found that the protein abundances of these 11 genes were remarkably upregulated in lung adenocarcinomas compared with normal tissues. In conclusion, by the WGCNA method, a panel of 11 genes were identified as predictive biomarkers for tumorigenesis and poor prognosis of lung adenocarcinomas.
Collapse
Affiliation(s)
- Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianye Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anping Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
37
|
Meng J, Gao L, Zhang M, Gao S, Fan S, Liang C. Systematic investigation of the prognostic value of cell division cycle-associated proteins for clear cell renal cell carcinoma patients. Biomark Med 2020; 14:223-238. [PMID: 31955607 DOI: 10.2217/bmm-2019-0498] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: To explore the prognostic value of the cell division cycle-associated proteins (CDCA) family in clear cell renal cell carcinoma. Methods: Gene profiles were collected from the The Cancer Genome Atlas-Kidney Renal Clear Cell Carcinoma (TCGA-KIRC), the GSE29609 and GSE22541 datasets. Genetic alteration and DNA methylation data were downloaded from the cBioPortal and MethSurv. The functional enrichment data were analyzed by Metascape. Results: The mRNA expression of the CDCAs, except CBX2, was significantly increased in clear cell renal cell carcinoma patients. Genetic alterations might affect the expression of CDCAs, but promotor methylation does not affect CDCA gene expression. The overall expression of the CDCAs, according to the The Cancer Genome Atlas-KIRC database (hazard ratio [HR]: 2.18), the GSE29609 (HR: 6.08) and GSE22541 (HR: 6.73), was significantly associated with unfavorable overall survival. In addition, genes co-expressed with CDCAs (R2 ≥0.3) were highly associated with cell division and the FOXM1 pathway. Conclusion: Our study demonstrated that the aberrant expression of CDCA gene family members plays an indispensable role in tumorigenesis.
Collapse
Affiliation(s)
- Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology & Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Lei Gao
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, PR China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology & Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Shenglin Gao
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, P.R. China
| | - Song Fan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology & Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University; Institute of Urology & Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
38
|
Wan Z, Zhang X, Luo Y, Zhao B. Identification of Hepatocellular Carcinoma-Related Potential Genes and Pathways Through Bioinformatic-Based Analyses. Genet Test Mol Biomarkers 2019; 23:766-777. [PMID: 31633428 DOI: 10.1089/gtmb.2019.0063] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Zheng Wan
- School of Medicine, Xiamen University, Xiamen, China
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | | | - Yuyang Luo
- ShenZhen College of International Education, Shenzhen, China
| | - Bin Zhao
- School of Medicine, Xiamen University, Xiamen, China
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| |
Collapse
|