1
|
Shu Y, Pang X, Li H, Deng C. A multidimensional analysis of ZW10 interacting kinetochore protein in human tumors. Am J Cancer Res 2024; 14:390-402. [PMID: 38323280 PMCID: PMC10839319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
ZW10 interacting kinetochore protein (ZWINT), an essential part of the kinetochore complex, plays a crucial role in maintaining genome stability by correcting improper attachments between the kinetochore and microtubules. An initial analysis of The Cancer Genome Atlas and Gene Expression Omnibus databases revealed that ZWINT is significantly expressed across a diverse range of tumor types. We subsequently investigated the influence of ZWINT on clinical outcomes and potential signaling pathways. A multidimensional analysis of ZWINT revealed significant statistical associations between ZWINT expression and clinical outcomes, as well as the E2F1 oncogenic signature. Experimental validation confirmed the increased expression of ZWINT in both pancreatic cancer cell lines and pancreatic adenocarcinoma tissues. Furthermore, our findings indicate that ZWINT promotes the proliferation of PANC-1 cells through cell cycle regulation. This comprehensive analysis of ZWINT suggests a strong correlation between its expression and various types of tumors, especially pancreatic adenocarcinoma (PAAD), indicating its potential oncogenic role. These findings enhance our understanding of the function of ZWINT in carcinogenesis.
Collapse
Affiliation(s)
- Yufeng Shu
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Xiaoyang Pang
- Department of Spinal Surgery, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Chao Deng
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| |
Collapse
|
2
|
Juárez-Barber E, Corachán A, Carbajo-García MC, Faus A, Vidal C, Giles J, Pellicer A, Cervelló I, Ferrero H. Transcriptome analysis of adenomyosis eutopic endometrium reveals molecular mechanisms involved in adenomyosis-related implantation failure and pregnancy disorders. Reprod Biol Endocrinol 2024; 22:10. [PMID: 38195505 PMCID: PMC10775471 DOI: 10.1186/s12958-023-01182-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Women with adenomyosis are characterized by having defective decidualization, impaired endometrial receptivity and/or embryo-maternal communication, and implantation failure. However, the molecular mechanisms underlying adenomyosis-related infertility remain unknown, mainly because of the restricted accessibility and the difficult preservation of endometrial tissue in vitro. We have recently shown that adenomyosis patient-derived endometrial organoids, maintain disease-specific features while differentiated into mid-secretory and gestational endometrial phase, overcoming these research barriers and providing a robust platform to study adenomyosis pathogenesis and the associated molecular dysregulation related to implantation and pregnancy disorders. For this reason, we aim to characterize the dysregulated mechanisms in the mid-secretory and gestational endometrium of patients with adenomyosis by RNA-sequencing. METHODS Endometrial organoids were derived from endometrial biopsies collected in the proliferative phase of women with adenomyosis (ADENO) or healthy oocyte donors (CONTROL) (n = 15/group) and differentiated into mid-secretory (-SECorg) and gestational (-GESTorg) phases in vitro. Following RNA-sequencing, the significantly differentially expressed genes (DEGs) (FDR < 0.05) were identified and selected for subsequent functional enrichment analysis and QIAGEN Ingenuity Pathway Analysis (IPA). Statistical differences in gene expression were evaluated with the Student's t-test or Wilcoxon test. RESULTS We identified 1,430 DEGs in ADENO-SECorg and 1,999 DEGs in ADENO-GESTorg. In ADENO-SECorg, upregulated genes included OLFM1, FXYD5, and RUNX2, which are involved in impaired endometrial receptivity and implantation failure, while downregulated genes included RRM2, SOSTDC1, and CHAC2 implicated in recurrent implantation failure. In ADENO-GESTorg, upregulated CXCL14 and CYP24A1 and downregulated PGR were related to pregnancy loss. IPA predicted a significant inhibition of ID1 signaling, histamine degradation, and activation of HMGB1 and Senescence pathways, which are related to implantation failure. Alternatively, IPA predicted an inhibition of D-myo-inositol biosynthesis and VEGF signaling, and upregulation of Rho pathway, which are related to pregnancy loss and preeclampsia. CONCLUSIONS Identifying dysregulated molecular mechanisms in mid-secretory and gestational endometrium of adenomyosis women contributes to the understanding of adenomyosis-related implantation failure and/or pregnancy disorders revealing potential therapeutic targets. Following experimental validation of our transcriptomic and in silico findings, our differentiated adenomyosis patient-derived organoids have the potential to provide a reliable platform for drug discovery, development, and personalized drug screening for affected patients.
Collapse
Affiliation(s)
- Elena Juárez-Barber
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
| | - Ana Corachán
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
- Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, Valencia, 46010, Spain
| | - María Cristina Carbajo-García
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
- Department of Pediatrics, Obstetrics and Gynecology, Universidad de Valencia, Valencia, 46010, Spain
| | - Amparo Faus
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
| | | | - Juan Giles
- IVI-RMA Valencia, Valencia, 46015, Spain
| | - Antonio Pellicer
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
- IVI-RMA Rome, Rome, 00197, Italy
| | - Irene Cervelló
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain
| | - Hortensia Ferrero
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Valencia, 46026, Spain.
| |
Collapse
|
3
|
Bai L, Xiang Y, Tang M, Liu S, Chen Q, Chen Q, Zhang M, Wan S, Sang Y, Li Q, Wang S, Li Z, Song Y, Hu X, Mao L, Feng G, Cui L, Ye Y, Zhu Y. ALKBH5 controls the meiosis-coupled mRNA clearance in oocytes by removing the N 6-methyladenosine methylation. Nat Commun 2023; 14:6532. [PMID: 37848452 PMCID: PMC10582257 DOI: 10.1038/s41467-023-42302-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 10/06/2023] [Indexed: 10/19/2023] Open
Abstract
N6-methyladenosine (m6A) maintains maternal RNA stability in oocytes. One regulator of m6A, ALKBH5, reverses m6A deposition and is essential in RNA metabolism. However, the specific role of ALKBH5 in oocyte maturation remains elusive. Here, we show that Alkbh5 depletion causes a wide range of defects in oocyte meiosis and results in female infertility. Temporal profiling of the maternal transcriptomes revealed striking RNA accumulation in Alkbh5-/- oocytes during meiotic maturation. Analysis of m6A dynamics demonstrated that ALKBH5-mediated m6A demethylation ensures the timely degradation of maternal RNAs, which is severely disrupted following Alkbh5-/- depletion. A distinct subset of transcripts with persistent m6A peaks are recognized by the m6A reader IGF2BP2 and thus remain stabilized, resulting in impaired RNA clearance. Additionally, reducing IGF2BP2 in Alkbh5-depleted oocytes partially rescued these defects. Overall, this work identifies ALKBH5 as a key determinant of oocyte quality and unveil the facilitating role of ALKBH5-mediated m6A removal in maternal RNA decay.
Collapse
Affiliation(s)
- Long Bai
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
| | - Yu Xiang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Minyue Tang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Shuangying Liu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Qingqing Chen
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Qichao Chen
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Min Zhang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Shan Wan
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yimiao Sang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Qingfang Li
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Sisi Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Zhekun Li
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yang Song
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Xiaoling Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Luna Mao
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Guofang Feng
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Long Cui
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yinghui Ye
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yimin Zhu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310006, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
- Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China.
| |
Collapse
|
4
|
Ma Z, Cai Y, Tian C. ZWINT promotes the proliferation, migration, and invasion of cervical cancer cells by regulating the p53/p21 signaling pathway. CHINESE J PHYSIOL 2023; 66:372-378. [PMID: 37929349 DOI: 10.4103/cjop.cjop-d-23-00001] [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] [Indexed: 11/07/2023] Open
Abstract
Cervical cancer leads to 300,000 deaths annually and the mechanism of cervical carcinogenesis remains unclear. Zeste White 10-interacting kinetochore protein (ZWINT) is uniquely elevated in malignancies, promoting proliferation, migration, and colony formation of cancer cells. To investigate the role of ZWINT in proliferation, migration, invasion of cervical cancer, and evaluate the potential ability of ZWINT as a therapeutic target. First, ZWINT expression in cervical cancer was analyzed using the bioinformatic methods and assessed in several cervical cancer cell lines. The cell viability and colony formation assays were used to evaluate cell proliferation. Then, transwell assay was performed to investigate cell migration and invasion. Moreover, western blot was used to measure the expression level of ZWINT, matrix metalloproteinase 9 (MMP-9), N-cadherin, E-cadherin, p53 and p21 in CaSki and HeLa cells with ZWINT overexpression or knockdown. The bioinformatic analysis and western blot assay revealed the expression of ZWINT was significantly increased in cervical cancer. The cell viability and colony formation analysis illustrated that cell proliferation could be promoted by ZWINT overexpression and suppressed by ZWINT knockdown. Moreover, ZWINT promoted migration and invasion of CaSki and HeLa cells, through regulating the expression of MMP-9, N-cadherin, and E-cadherin. Furthermore, ZWINT attenuated the expression of p53 and p21 in cervical cancer cells. In summary, ZWINT functions in promoting cell proliferation, migration, and invasion of cervical cancer cells by suppressing p53/p21 signaling pathway, which indicated ZWINT is a potential therapeutic target for cervical cancer treatment.
Collapse
Affiliation(s)
- Zhe Ma
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Beihua University, Jilin, Jilin Province, China
| | - Yufei Cai
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Beihua University, Jilin, Jilin Province, China
| | - Chenchen Tian
- Department of Gynaecology and Obstetrics, Affiliated Hospital of Beihua University, Jilin, Jilin Province, China
| |
Collapse
|
5
|
Cao S, Xiao S, Zhang J, Li S. Identification of the cell cycle characteristics of non-small cell lung cancer and its relationship with tumor immune microenvironment, cell death pathways, and metabolic reprogramming. Front Endocrinol (Lausanne) 2023; 14:1147366. [PMID: 37091844 PMCID: PMC10117961 DOI: 10.3389/fendo.2023.1147366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/08/2023] [Indexed: 04/09/2023] Open
Abstract
BackgroundThe genes related to the cell cycle progression could be considered the key factors in human cancers. However, the genes involved in cell cycle regulation in non-small cell lung cancer (NSCLC) have not yet been reported. Therefore, it is necessary to evaluate the genes related to the cell cycle in all types of cancers, especially NSCLC.MethodsThis study constituted the first pan-cancer landscape of cell cycle signaling. Cluster analysis based on cell cycle signaling was conducted to identify the potential molecular heterogeneity of NSCLC. Further, the discrepancies in the tumor immune microenvironment, metabolic remodeling, and cell death among the three clusters were investigated. Immunohistochemistry was performed to validate the protein levels of the ZWINT gene and examine its relationship with the clinical characteristics. Bioinformatics analyses and experimental validation of the ZWINT gene were also conducted.ResultsFirst, pan-cancer analysis provided an overview of cell cycle signaling and highlighted its crucial role in cancer. A majority of cell cycle regulators play risk roles in lung adenocarcinoma (LUAD); however, some cell cycle genes play protective roles in lung squamous cell carcinoma (LUSC). Cluster analysis revealed three potential subtypes for patients with NSCLC. LUAD patients with high cell cycle activities were associated with worse prognosis; while, LUSC patients with high cell cycle activities were associated with a longer survival time. Moreover, the above three subtypes of NSCLC exhibited distinct immune microenvironments, metabolic remodeling, and cell death pathways. ZWINT, a member of the cell signaling pathway, was observed to be significantly associated with the prognosis of LUAD patients. A series of experiments verified the higher expression levels of ZWINT in NSCLC compared to those in paracancerous tissues. The activation of epithelial-mesenchymal transition (EMT) induced by ZWINT might be responsible for tumor progression.ConclusionThis study revealed the regulatory function of the cell cycle genes in NSCLC, and the molecular classification based on cell cycle-associated genes could evaluate the different prognoses of patients with NSCLC. ZWINT expression was found to be significantly upregulated in NSCLC tissues, which might promote tumor progression via activation of the EMT pathway.
Collapse
Affiliation(s)
- Shengji Cao
- Department of Clinical Laboratory Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Sitong Xiao
- Department of Clinical Laboratory Medicine, The Third People’s Hospital of Yuhang District, Hangzhou, China
| | - Jingyang Zhang
- Department of Clinical Laboratory Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shijun Li
- Department of Clinical Laboratory Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Shijun Li,
| |
Collapse
|
6
|
Identification of Human Cell Cycle Phase Markers Based on Single-Cell RNA-Seq Data by Using Machine Learning Methods. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2516653. [PMID: 36004205 PMCID: PMC9393965 DOI: 10.1155/2022/2516653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 12/17/2022]
Abstract
The cell cycle is composed of a series of ordered, highly regulated processes through which a cell grows and duplicates its genome and eventually divides into two daughter cells. According to the complex changes in cell structure and biosynthesis, the cell cycle is divided into four phases: gap 1 (G1), DNA synthesis (S), gap 2 (G2), and mitosis (M). Determining which cell cycle phases a cell is in is critical to the research of cancer development and pharmacy for targeting cell cycle. However, current detection methods have the following problems: (1) they are complicated and time consuming to perform, and (2) they cannot detect the cell cycle on a large scale. Rapid developments in single-cell technology have made dissecting cells on a large scale possible with unprecedented resolution. In the present research, we construct efficient classifiers and identify essential gene biomarkers based on single-cell RNA sequencing data through Boruta and three feature ranking algorithms (e.g., mRMR, MCFS, and SHAP by LightGBM) by utilizing four advanced classification algorithms. Meanwhile, we mine a series of classification rules that can distinguish different cell cycle phases. Collectively, we have provided a novel method for determining the cell cycle and identified new potential cell cycle-related genes, thereby contributing to the understanding of the processes that regulate the cell cycle.
Collapse
|
7
|
Liu X, Wu H, Liu Z. An Integrative Human Pan-Cancer Analysis of Cyclin-Dependent Kinase 1 (CDK1). Cancers (Basel) 2022; 14:cancers14112658. [PMID: 35681641 PMCID: PMC9179585 DOI: 10.3390/cancers14112658] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Cyclin-dependent kinase 1 (CDK1), one of the key regulators of the G2/M checkpoint, is expressed in many cells and plays an important role in cell cycle control. However, CDK1 expression is substantially increased in many tumors of diverse origins and is associated with tumorigenesis. Targeting CDK1 shows promising results for several tumors. However, a systematic and integrative analysis of CDK1 in cancer has not been conducted. The present study aims to use pan-cancer analysis to investigate the relationship, similarities, and differences in genetic and cellular changes associated with CDK1 in various tumors and tumor microenvironments. Our findings elucidate that CDK1 expression increases in more than 20 human tumors and is highly correlated with oncogenic signature gene sets, biological pathways, immune cell infiltration, tumor mutational burden, microsatellite instability, and lower survival rate across multiple tumors. Targeting CDK1 may provide a novel and effective strategy for cancer immunotherapy. Abstract Cyclin-dependent kinase 1 (CDK1) is essential for cell division by regulating the G2/M phase and mitosis. CDK1 overexpression can also promote the development and progression of a variety of cancers. However, the significance of CDK1 in the formation, progression, and prognosis of human pan-cancer remains unclear. In the present study, we used The Cancer Genome Atlas database, Clinical Proteomic Tumor Analysis Consortium, Human Protein Atlas, Genotype-Tissue Expression, and other well-established databases to comprehensively examine CDK1 genetic alterations and gene/protein expression in various cancers and their relationships with the prognosis, immune reactivities, and clinical outcomes for 33 tumor types. Gene set enrichment analysis was also conducted to examine the potential mechanisms of CDK1 in tumorigenesis. The data showed that CDK1 mutation was frequently present in multiple tumors. CDK1 expression was significantly increased in various types of tumors as compared with normal tissues and was associated with poor overall and disease-free survival. In addition, CDK1 expression was significantly correlated with oncogenic genes, proteins, cellular components, myeloid-derived suppressor cell infiltration, ESTMATEScore, and signaling pathways associated with tumor development and progression and tumor microenvironments. These data indicate that CDK1 could serve as a promising biomarker for predicting tumor prognosis and a potential target for cancer treatment.
Collapse
Affiliation(s)
- Xuanyou Liu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA;
| | - Hao Wu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA;
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Correspondence: ; Tel.: +573-884-3278
| |
Collapse
|
8
|
Xie C, Lu C, Liu Y, Liu Z. Diagnostic gene biomarkers for predicting immune infiltration in endometriosis. BMC Womens Health 2022; 22:184. [PMID: 35585523 PMCID: PMC9118874 DOI: 10.1186/s12905-022-01765-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/06/2022] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine the potential diagnostic markers and extent of immune cell infiltration in endometriosis (EMS). METHODS Two published profiles (GSE7305 and GSE25628 datasets) were downloaded, and the candidate biomarkers were identified by support vector machine recursive feature elimination analysis and a Lasso regression model. The diagnostic value and expression levels of biomarkers in EMS were verified by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting, then further validated in the GSE5108 dataset. CIBERSORT was used to estimate the composition pattern of immune cell components in EMS. RESULTS One hundred and fifty-three differential expression genes (DEGs) were identified between EMS and endometrial with 83 upregulated and 51 downregulated genes. Gene sets related to arachidonic acid metabolism, cytokine-cytokine receptor interactions, complement and coagulation cascades, chemokine signaling pathways, and systemic lupus erythematosus were differentially activated in EMS compared with endometrial samples. Aquaporin 1 (AQP1) and ZW10 binding protein (ZWINT) were identified as diagnostic markers of EMS, which were verified using qRT-PCR and western blotting and validated in the GSE5108 dataset. Immune cell infiltrate analysis showed that AQP1 and ZWINT were correlated with M2 macrophages, NK cells, activated dendritic cells, T follicular helper cells, regulatory T cells, memory B cells, activated mast cells, and plasma cells. CONCLUSION AQP1 and ZWINT could be regarded as diagnostic markers of EMS and may provide a new direction for the study of EMS pathogenesis in the future.
Collapse
Affiliation(s)
- Chengmao Xie
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| | - Chang Lu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Yong Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Zhaohui Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| |
Collapse
|
9
|
Li W, Feng SS, Wu H, Deng J, Zhou WY, Jia MX, Shi Y, Ma L, Zeng XX, Zuberi Z, Fu D, Liu X, Chen Z. Comprehensive Analysis of CDK1-Associated ceRNA Network Revealing the Key Pathways LINC00460/LINC00525-Hsa-Mir-338-FAM111/ZWINT as Prognostic Biomarkers in Lung Adenocarcinoma Combined with Experiments. Cells 2022; 11:cells11071220. [PMID: 35406786 PMCID: PMC8997540 DOI: 10.3390/cells11071220] [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: 01/25/2022] [Revised: 03/20/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the leading cause of cancer deaths worldwide, and effective biomarkers are still lacking for early detection and prognosis prediction. Here, based on gene expression profiles of LUAD patients from The Cancer Genome Atlas (TCGA), 806 long non-coding RNAs (lncRNAs), 122 microRNAs (miRNAs) and 1269 mRNAs associated with CDK1 were identified. The regulatory axis of LINC00460/LINC00525-hsa-mir-338-FAM111B/ZWINT was determined according to the correlation between gene expression and patient prognosis. The abnormal up-regulation of FAM111B/ZWINT in LUAD was related to hypomethylation. Furthermore, immune infiltration analysis suggested FAM111B/ZWINT could affect the development and prognosis of cancer by regulating the LUAD immune microenvironment. EMT feature analysis suggested that FAM111B/ZWINT promoted tumor spread through the EMT process. Functional analysis showed FAM111B/ZWINT was involved in cell cycle events such as DNA replication and chromosome separation. We analyzed the HERB and GSCALite databases to identify potential target medicines that may play a role in the treatment of LUAD. Finally, the expression of LINC00460/LINC00525-hsa-mir-338-FAM111B/ZWINT axis was verified in LUAD cells by RT-qPCR, and these results were consistent with bioinformatics analysis. Overall, we constructed a CDK1-related ceRNA network and revealed the LINC00460/LINC00525-hsa-mir-338-FAM111/ZWINT pathways as potential diagnostic biomarkers or therapeutic targets of LUAD.
Collapse
Affiliation(s)
- Wen Li
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Shan-Shan Feng
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Hao Wu
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Jing Deng
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Wang-Yan Zhou
- Department of Medical Record, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang 421001, China;
| | - Ming-Xi Jia
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Yi Shi
- National Engineering Research Center of Rice and Byproduct Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, China; (H.W.); (M.-X.J.); (Y.S.)
| | - Liang Ma
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Xiao-Xi Zeng
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
| | - Zavuga Zuberi
- Department of Science and Laboratory Technology, Dar es Salaam Institute of Technology, Dar es Salaam P.O. Box 2958, Tanzania;
| | - Da Fu
- Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China;
| | - Xiang Liu
- Department of Thoracic Surgery, Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang 421001, China
- Correspondence: (X.L.); (Z.C.); Tel.: +86-0734-889-9990 (X.L.); +86-158-6971-6968 (Z.C.)
| | - Zhu Chen
- College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou 412007, China; (W.L.); (S.-S.F.); (J.D.); (L.M.); (X.-X.Z.)
- Correspondence: (X.L.); (Z.C.); Tel.: +86-0734-889-9990 (X.L.); +86-158-6971-6968 (Z.C.)
| |
Collapse
|
10
|
Yue W, Wang Y, Meng T, Zhang H, Zhang X, Ouyang Y, Hou Y, Schatten H, Wang Z, Sun Q. Kinetochore scaffold 1 regulates SAC function during mouse oocyte meiotic maturation. FASEB J 2022; 36:e22210. [DOI: 10.1096/fj.202101586rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
- University of Chinese Academy of Sciences Beijing China
| | - Yue Wang
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Tie‐Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Hong‐Yong Zhang
- Department of Reproductive Medicine Peking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical Center Shenzhen China
| | - Xin‐Ran Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
- University of Chinese Academy of Sciences Beijing China
| | - Ying‐Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Heide Schatten
- Department of Veterinary Pathobiology University of Missouri Columbia Missouri USA
| | - Zhen‐Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
- University of Chinese Academy of Sciences Beijing China
| | - Qing‐Yuan Sun
- Fertility Preservation Lab, Guangdong‐Hong Kong Metabolism & Reproduction Joint Laboratory Reproductive Medicine Center Guangdong Second Provincial General Hospital Guangzhou China
| |
Collapse
|
11
|
Roy B, Sim J, Han SJY, Joglekar AP. Kre28-Spc105 interaction is essential for Spc105 loading at the kinetochore. Open Biol 2022; 12:210274. [PMID: 35042402 PMCID: PMC8767186 DOI: 10.1098/rsob.210274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/18/2021] [Indexed: 11/12/2022] Open
Abstract
Kinetochore (KTs) are macromolecular protein assemblies that attach sister chromatids to spindle microtubules (MTs) and mediate accurate chromosome segregation during mitosis. The outer KT consists of the KMN network, a protein super-complex comprising Knl1 (yeast Spc105), Mis12 (yeast Mtw1), and Ndc80 (yeast Ndc80), which harbours sites for MT binding. Within the KMN network, Spc105 acts as an interaction hub of components involved in spindle assembly checkpoint (SAC) signalling. It is known that Spc105 forms a complex with KT component Kre28. However, where Kre28 physically localizes in the budding yeast KT is not clear. The exact function of Kre28 at the KT is also unknown. Here, we investigate how Spc105 and Kre28 interact and how they are organized within bioriented yeast KTs using genetics and cell biological experiments. Our microscopy data show that Spc105 and Kre28 localize at the KT with a 1 : 1 stoichiometry. We also show that the Kre28-Spc105 interaction is important for Spc105 protein turn-over and essential for their mutual recruitment at the KTs. We created several truncation mutants of kre28 that affect Spc105 loading at the KTs. When over-expressed, these mutants sustain the cell viability, but SAC signalling and KT biorientation are impaired. Therefore, we conclude that Kre28 contributes to chromosome biorientation and high-fidelity segregation at least indirectly by regulating Spc105 localization at the KTs.
Collapse
Affiliation(s)
- Babhrubahan Roy
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Janice Sim
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Simon J. Y. Han
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ajit P. Joglekar
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
12
|
Chen P, He Z, Wang J, Xu J, Jiang X, Chen Y, Liu X, Jiang J. Hypoxia-Induced ZWINT Mediates Pancreatic Cancer Proliferation by Interacting With p53/p21. Front Cell Dev Biol 2021; 9:682131. [PMID: 34900978 PMCID: PMC8652205 DOI: 10.3389/fcell.2021.682131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
p53/p21 signaling plays a vital role in pancreatic cancer (PC) progression. ZWINT was shown to function as an oncoprotein in the progression of multiple cancers. However, the involvement of ZWINT and p53 activation in the progression of PC remains poorly understood. Bioinformatics and tissue array chip analyses were performed to evaluate ZWINT expression in pancreatic cancer. ZWINT mRNA and protein expression were evaluated in normoxia and hypoxia. CHIP was used to evaluate HIF1α interaction with the ZWINT promoter. CCK8, colony formation, EDU, and cell cycle analysis were used to examine PC cell proliferation. Immunoprecipitation and immunofluorescence were used to examine the interaction of ZWINT, MDM2, and p53. p53 activity was evaluated by q-PCR and luciferase assay. Protein degradation and ubiquitination assays were used to analyze the role of ZWINT in p53 ubiquitination. ZWINT was overexpressed in pancreatic cancer and induced in hypoxia. ZWINT promoted pancreatic cancer growth and cell cycle progression. Bioinformatic analysis revealed that ZWINT may regulate the p53 signal pathway. ZWINT interacts with p53 and promotes its ubiquitination and degradation. ZWINT promoted proliferation via p53/p21. Immunohistochemistry of clinical specimens revealed that that ZWINT expression was significantly negatively correlated with p53/p21. Our data showed that hypoxia regulates the expression of ZWINT, which activated p53/p21 signaling pathway to promote PC growth.
Collapse
Affiliation(s)
- Peng Chen
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jie Wang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jian Xu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xueyi Jiang
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yankun Chen
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xinyuan Liu
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Lin T, Zhang Y, Lin Z, Peng L. ZWINT is a Promising Therapeutic Biomarker Associated with the Immune Microenvironment of Hepatocellular Carcinoma. Int J Gen Med 2021; 14:7487-7501. [PMID: 34744456 PMCID: PMC8566006 DOI: 10.2147/ijgm.s340057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
Background The prognosis of patients with advanced hepatocellular carcinoma (HCC) is still poor, effective therapeutic targets are needed. ZW10 interacting kinetochore protein (Zwint) is an essential component of the mitotic spindle checkpoint and is upregulated in cancers. Disappointing, the role of ZWINT in HCC has not been fully illuminated. Methods Multiple tools, including TIMER2.0, Oncomine, GEPIA2, UALCAN, LinkedOmics, Kaplan-Meier Plotter, cBioPortal, and MethSurv, etc. were applied to comprehensively analyze the expression, genetic alternations, clinicopathological relevance, prognostic value, and DNA methylation of ZWINT, along with its correlations with immune infiltration in HCC. Besides, gene set enrichment analysis (GSEA) and protein-protein interaction (PPI) analysis were performed for the correlated genes of ZWINT, closely interconnected clusters and hub proteins in the PPI network were discovered to learn the underlying biological mechanisms. Results We found ZWINT was significantly upregulated in diverse cancers including HCC, compared with the corresponding normal controls. ZWINT upregulation was significantly associated with unfavorable clinicopathological features and survivals of HCC patients. Genetic alternations of ZWINT frequently occurred, which were linked to worse outcomes of HCC patients. The results of GSEA displayed ZWINT and its correlated genes might be components of condensed chromosomes and spindles, which participated in biological processes and signaling pathways involving DNA replication, cytokinesis, and cell cycle checkpoint, etc. Three highly interconnected clusters and 10 hub proteins were identified from the PPI network constructed with the correlated genes of ZWINT. Moreover, ZWINT expression was found positively correlated with infiltration levels of various immune cells, especially myeloid-derived suppressor cells. Conclusion This study demonstrated ZWINT might be a promising unfavorable prognostic biomarker and a therapeutic target of HCC, which could regulate HCC progression through cell division and immunosuppression.
Collapse
Affiliation(s)
- Tong Lin
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, People's Republic of China
| | - Yingzhao Zhang
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, People's Republic of China
| | - Zhimei Lin
- The Fourth Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, People's Republic of China
| | - Lisheng Peng
- Department of Science and Education, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, People's Republic of China
| |
Collapse
|
14
|
Mou K, Zhang J, Mu X, Wang L, Liu W, Ge R. Zwint facilitates melanoma progression by promoting c-Myc expression. Exp Ther Med 2021; 22:818. [PMID: 34131441 PMCID: PMC8193213 DOI: 10.3892/etm.2021.10250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
ZW10 interactor (Zwint) is upregulated in various types of tumors and exerts a carcinogenic effect. However, little is known about the expression profile, function and molecular mechanisms of action of Zwint in melanoma. Therefore, the aim of the present study was to investigate the expression levels of Zwint in melanoma cell lines and tissues. It was revealed that Zwint was highly expressed in melanoma samples. Functional experiments indicated that Zwint knockdown suppressed the proliferation and migration of A375 melanoma cells. Further mechanistic studies demonstrated that Zwint knockdown decreased the protein expression levels of c-Myc, MMP-2, Slug, mTOR, phosphorylated (p)-mTOR, p-p38 and fibronectin, while it increased the protein expression levels of E-cadherin and MMP-9. Among these genes, c-Myc, MMP-2 and Slug were overexpressed to investigate their effects on cell proliferation following Zwint knockdown. The results demonstrated that overexpression of c-Myc, but not MMP-2 or Slug, rescued the effects of Zwint knockdown on melanoma cell proliferation and migration. Taken together, the results of the present study suggested that Zwint may act as an oncogene in melanoma by regulating c-Myc expression.
Collapse
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jian Zhang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Mu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lijuan Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenli Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Ge
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
15
|
Kim JH, Youn Y, Lee JC, Kim J, Hwang JH. Involvement of the NF-κB signaling pathway in proliferation and invasion inhibited by Zwint-1 deficiency in Pancreatic Cancer Cells. J Cancer 2020; 11:5601-5611. [PMID: 32913455 PMCID: PMC7477444 DOI: 10.7150/jca.46173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023] Open
Abstract
Pancreatic cancer (PC) is an intractable cancer that is difficult to diagnose early and has a 5-year survival rate of less than 8%. ZW10-interacting kinetochore protein (ZWINT) is a crucial gene that contributes to chromosome instability and is essential for spindle assembly and kinetochore-microtubule attachment during meiosis and mitosis. However, the mechanism through which Zwint-1 promotes PC progression is yet to be elucidated. Here, we report that Zwint-1 is highly expressed in clinical PC specimens (based on analysis of the Gene Expression Profiling Interactive Analysis database) and various PC cell lines. Importantly, Zwint-1-deficient PC cells showed reduced nuclear factor-kappa B (NF-κB) (Ser536) phosphorylation along with inhibited proliferation and colony formation due to downregulation of NF-κB-regulated genes such as CCND1, cIAP1/2, and XIAP. In addition, Zwint-1-deficient PC cells showed reduced invasion and migration abilities, and decreased expression levels of the metalloproteinases MMP2 and MMP9. Furthermore, Zwint-1 deficiency arrested the PC cell cycle at the G2/M phase because the chromosomes failed to segregate properly, and the apoptosis rate in these cells gradually increased, accompanied by increased caspase-3 activation and anti-poly (ADP ribose) polymerase cleavage. Apoptosis caused by Zwint-1 deficiency was demonstrated to occur through caspase-dependent pathways based on experiments involving treatment with a pan-caspase inhibitor (Z-VAD-Fmk). Thus, Zwint-1 contributes to cell growth, invasion, and survival through NF-κB signaling pathways, suggesting that it could serve as a PC biomarker and new therapeutic target.
Collapse
Affiliation(s)
- Jae Hyeong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Yuna Youn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jong-chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| |
Collapse
|
16
|
Kixmoeller K, Allu PK, Black BE. The centromere comes into focus: from CENP-A nucleosomes to kinetochore connections with the spindle. Open Biol 2020; 10:200051. [PMID: 32516549 PMCID: PMC7333888 DOI: 10.1098/rsob.200051] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic chromosome segregation relies upon specific connections from DNA to the microtubule-based spindle that forms at cell division. The chromosomal locus that directs this process is the centromere, where a structure called the kinetochore forms upon entry into mitosis. Recent crystallography and single-particle electron microscopy have provided unprecedented high-resolution views of the molecular complexes involved in this process. The centromere is epigenetically specified by nucleosomes harbouring a histone H3 variant, CENP-A, and we review recent progress on how it differentiates centromeric chromatin from the rest of the chromosome, the biochemical pathway that mediates its assembly and how two non-histone components of the centromere specifically recognize CENP-A nucleosomes. The core centromeric nucleosome complex (CCNC) is required to recruit a 16-subunit complex termed the constitutive centromere associated network (CCAN), and we highlight recent structures reported of the budding yeast CCAN. Finally, the structures of multiple modular sub-complexes of the kinetochore have been solved at near-atomic resolution, providing insight into how connections are made to the CCAN on one end and to the spindle microtubules on the other. One can now build molecular models from the DNA through to the physical connections to microtubules.
Collapse
Affiliation(s)
- Kathryn Kixmoeller
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Graduate Program in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Praveen Kumar Allu
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ben E Black
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Graduate Program in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
17
|
He Y, Li R, Gu L, Deng H, Zhao Y, Guo Y, Yu S, Wang G. Anaphase-promoting complex/cyclosome-Cdc-20 promotes Zwint-1 degradation. Cell Biochem Funct 2020; 38:451-459. [PMID: 31945194 DOI: 10.1002/cbf.3499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 02/05/2023]
Abstract
ZW10 interactor (Zwint-1) is an important component of the centromere and can recruit the dynamic protein kinase and dynein to promote chromosome movement and regulate the spindle assembly checkpoint (SAC). Zwint-1 activity is tightly regulated during the cell cycle. However, how the stability of Zwint-1 is regulated has not been clarified. Here, we show that the relative levels of Zwint-1 expression gradually decreased with the progression of cell cycling and decline sharply during mitotic exit. Treatment with cycloheximide reduced the levels of Zwint-1 while treatment with MG132 to inhibit endogenous ubiquitin-proteasome elevated the levels of Zwint-1 in HEK293T cells or Hela cells. Such data suggest that Zwint-1 may be degraded by endogenous ubiquitin-proteasome. Furthermore, induction of cell-division cycle protein 20 (Cdc20) overexpression decreased the levels of Zwint-1, which was abrogated by MG132 treatment. In contrast, Cdc20 silencing promoted the accumulation of Zwint-1. in vivo ubiquitination assay revealed that Cdc20 promoted the formation of Zwint-1 and ubiquitin-proteasome conjugates. Cotransfection with Cdc20 and wild-type Zwint-1, but not Zwint-1ΔD-box , reduced the levels of Zwint-1. Immunoprecipitation and western blot analyses showed that Cdc20 interacted with wild-type Zwint-1, but not Zwint-1ΔD-box although both Zwint-1 and Zwint-1ΔD-box overexpression did not induce mitotic arrest. Collectively, our data indicated that Zwint-1 was ubiquitinated by anaphase-promoting complex/cyclosome (APC/C)-Cdc20 in a D-box-dependent manner. Therefore, the APC/C-Cdc20 controls the stability of Zwint-1, ensuring accurate regulation of the spindle assembly during the cell cycling in HEK293T cells.
Collapse
Affiliation(s)
- Yan He
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Rui Li
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Liming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Huixiong Deng
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Yingzhu Guo
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Shun Yu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Gefei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Pathogen Biology and Immunology Center, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
18
|
A tumor-specific modulation of heterogeneous ribonucleoprotein A0 promotes excessive mitosis and growth in colorectal cancer cells. Cell Death Dis 2020; 11:245. [PMID: 32303675 PMCID: PMC7165183 DOI: 10.1038/s41419-020-2439-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 11/11/2022]
Abstract
RNA regulation mediating RNA-binding proteins (RBPs) have been shown to be related to the maintenance of homeostasis as well as cancer progression. However, the tumor-associated functions as well as the detailed mechanisms underlying the anti-tumor effects of most RBPs have yet to be explored. We herein report that the phosphorylated heterogeneous ribonucleoprotein (hnRNP) A0 promotes mitosis through the RAS-associated protein 3 GTPase-activating protein catalytic subunit 1 (RAB3GAP1)-Zeste white 10 interactor (ZWINT1) cascade. The downregulation assay of 20 representative hnRNPs, a major family of RNA-binding proteins, in colorectal cancer cells revealed that hnRNPA0 is a strong regulator of cancer cell growth. The tumor promotive function of hnRNPA0 was confirmed in gastrointestinal cancer cells, including pancreatic, esophageal, and gastric cancer cells, but not in non-cancerous cells. Flow cytometry and Western blotting analyses revealed that hnRNPA0 inhibited the apoptosis through the maintenance of G2/M phase promotion in colorectal cancer cells. A comprehensive analysis of mRNAs regulated by hnRNP A0 and immunostaining revealed that mitotic events were regulated by the hnRNPA0-RAB3GAP1 mRNA-mediated ZWINT-1 stabilization in colorectal cancer cells, but not in non-tumorous cells. The interaction of hnRNP A0 with mRNAs was dramatically changed by the deactivation of its phosphorylation site in cancer cells, but not in non-tumorous cells. Therefore, the tumor-specific biological functions characterized by the abnormal phosphorylation of RBPs are considered to be an attractive target for tumor treatment.
Collapse
|
19
|
Braun R, Anthuber L, Hirsch D, Wangsa D, Lack J, McNeil NE, Heselmeyer-Haddad K, Torres I, Wangsa D, Brown MA, Tubbs A, Auslander N, Gertz EM, Brauer PR, Cam MC, Sackett DL, Habermann JK, Nussenzweig A, Ruppin E, Zhang Z, Rosenberg DW, Ried T. Single-Cell-Derived Primary Rectal Carcinoma Cell Lines Reflect Intratumor Heterogeneity Associated with Treatment Response. Clin Cancer Res 2020; 26:3468-3480. [PMID: 32253233 DOI: 10.1158/1078-0432.ccr-19-1984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 01/22/2020] [Accepted: 04/01/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The standard treatment of patients with locally advanced rectal cancer consists of preoperative chemoradiotherapy (CRT) followed by surgery. However, the response of individual tumors to CRT is extremely diverse, presenting a clinical dilemma. This broad variability in treatment response is likely attributable to intratumor heterogeneity (ITH). EXPERIMENTAL DESIGN We addressed the impact of ITH on response to CRT by establishing single-cell-derived cell lines (SCDCL) from a treatment-naïve rectal cancer biopsy after xenografting. RESULTS Individual SCDCLs derived from the same tumor responded profoundly different to CRT in vitro. Clonal reconstruction of the tumor and derived cell lines based on whole-exome sequencing revealed nine separate clusters with distinct proportions in the SCDCLs. Missense mutations in SV2A and ZWINT were clonal in the resistant SCDCL, but not detected in the sensitive SCDCL. Single-cell genetic analysis by multiplex FISH revealed the expansion of a clone with a loss of PIK3CA in the resistant SCDCL. Gene expression profiling by tRNA-sequencing identified the activation of the Wnt, Akt, and Hedgehog signaling pathways in the resistant SCDCLs. Wnt pathway activation in the resistant SCDCLs was confirmed using a reporter assay. CONCLUSIONS Our model system of patient-derived SCDCLs provides evidence for the critical role of ITH for treatment response in patients with rectal cancer and shows that distinct genetic aberration profiles are associated with treatment response. We identified specific pathways as the molecular basis of treatment response of individual clones, which could be targeted in resistant subclones of a heterogenous tumor.
Collapse
Affiliation(s)
- Rüdiger Braun
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Lena Anthuber
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Daniela Hirsch
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Darawalee Wangsa
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland.,Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Nicole E McNeil
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | | | - Irianna Torres
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Danny Wangsa
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Markus A Brown
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Anthony Tubbs
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Noam Auslander
- Cancer Data Science Laboratory, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - E Michael Gertz
- Cancer Data Science Laboratory, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Philip R Brauer
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Margaret C Cam
- Office of Science and Technology Resources, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Dan L Sackett
- Eunice Kennedy Shriver National Institute of Child Health & Human Development, NIH, Bethesda, Maryland
| | - Jens K Habermann
- Section of Translational Surgical Oncology and Biobanking, Department of Surgery, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Germany
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Zhongqiu Zhang
- Department of Surgery, Waterbury Hospital, University of Connecticut School of Medicine, Waterbury, Connecticut
| | - Daniel W Rosenberg
- Center for Molecular Oncology, University of Connecticut Health, Farmington, Waterbury, Connecticut
| | - Thomas Ried
- Section of Cancer Genomics, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
20
|
Yang L, Han N, Zhang X, Zhou Y, Chen R, Zhang M. ZWINT: A potential therapeutic biomarker in patients with glioblastoma correlates with cell proliferation and invasion. Oncol Rep 2020; 43:1831-1844. [PMID: 32323832 PMCID: PMC7160549 DOI: 10.3892/or.2020.7573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary intracranial tumor in adults. Chemoradiotherapy resistance and recurrence after surgery are the main malignant progression factors, leading to a high mortality rate. Therefore, the exploration of novel biomarkers and molecular mechanisms of GBM is urgent. Differentially expressed genes (DEGs) of GBM were screened in a TCGA dataset. Homo sapiens ZW10 interacting kinetochore protein (ZWINT) was found to be upregulated in GBM, which was confirmed by immunohistochemical staining of a tissue microarray. Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) database. A protein-protein interaction (PPI) network was established by the STRING database, and hub genes were visualized by Cytoscape. The correlation results were verified with the GSE15824 dataset. Bioinformatic analysis confirmed that ZWINT was significantly positively correlated with kinetochore protein NDC80 homolog (NDC80), serine/threonine-protein kinase PLK1 (PLK1) and spindle and kinetochore associated complex subunit 1 (SKA1) and together are involved in regulating mitosis and the cell cycle of GBM. ZWINT expression was knocked down in U251 and U87 MG GBM cells by lentiviral vectors carrying a small hairpin RNA (shRNA) targeting ZWINT. The effect of ZWINT silencing on cell proliferation, invasion and apoptosis was determined by the Celigo assay, MTT assay, Transwell assay, flow cytometry and caspase-3/7 assay in vitro. A subcutaneous xenograft tumor model was established to explore the influence of ZWINT knockdown on GBM growth in vivo. Our preliminary study demonstrated that ZWINT knockdown effectively inhibited proliferation and invasion and induced apoptosis of GBM cells and notably suppressed GBM growth in vivo. Therefore, we speculate that ZWINT may be a potential therapeutic biomarker for GBM, with NDC80 and PLK1 conjointly involved in regulating cell division and the mitotic cell cycle.
Collapse
Affiliation(s)
- Li Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Na Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaoxi Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yangmei Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Rui Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Mengxian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
21
|
Sui L, Zhang S, Huang R, Li Z. HDAC11 promotes meiotic apparatus assembly during mouse oocyte maturation via decreasing H4K16 and α-tubulin acetylation. Cell Cycle 2020; 19:354-362. [PMID: 31910069 DOI: 10.1080/15384101.2019.1711315] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The smallest histone deacetylase (HDAC) and the solely member of class IV, HDAC11, is reported to regulate mitosis process and tumorigenesis, yet its roles in meiosis process remain unknown. In the present study, we first analyzed the expression of HDAC11 in mouse oocytes. HDAC11 showed gradual lower expression from GV (Germinal Vesicle) to MII (Metaphase II) stage oocytes. Then, the specific inhibitor of HDAC11, JB3-22 was used to explore the role of HDAC11 during mouse oocytes maturation. We found that inhibition of HDAC11 significantly interrupted mouse oocytes meiosis progress, caused abnormal spindle organization and misaligned chromosomes, impaired kinetochore-microtubule attachment and spindle assembly checkpoint (SAC) function. Moreover, HDAC11 inhibition significantly increased the acetylation level of α-tubulin that is associated with microtubule stability, and increased acetylation level of H4K16 that is important for kinetochore function. In conclusion, our study indicates that HDAC11 is an essential factor for oocytes maturation and it promotes meiotic process most likely though decreasing acetylation status of α-tubulin and H4K16.
Collapse
Affiliation(s)
- Liyan Sui
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, Jilin, China
| | - Sheng Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, Jilin, China
| | - Rong Huang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, Jilin, China
| | - Ziyi Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Liu J, Meng H, Li S, Shen Y, Wang H, Shan W, Qiu J, Zhang J, Cheng W. Identification of Potential Biomarkers in Association With Progression and Prognosis in Epithelial Ovarian Cancer by Integrated Bioinformatics Analysis. Front Genet 2019; 10:1031. [PMID: 31708970 PMCID: PMC6822059 DOI: 10.3389/fgene.2019.01031] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/25/2019] [Indexed: 02/03/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the malignancies in women, which has the highest mortality. However, the microlevel mechanism has not been discussed in detail. The expression profiles GSE27651, GSE38666, GSE40595, and GSE66957 including 188 tumor and 52 nontumor samples were downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) were filtered using R software, and we performed functional analysis using the clusterProfiler. Cytoscape software, the molecular complex detection plugin and database STRING analyzed DEGs to construct protein-protein interaction network. We identified 116 DEGs including 81 upregulated and 35 downregulated DEGs. Functional analysis revealed that they were significantly enriched in the extracellular region and biosynthesis of amino acids. We next identified four bioactive compounds (vorinostat, LY-294002,trichostatin A, and tanespimycin) based on ConnectivityMap. Then 114 nodes were obtained in protein-protein interaction. The three most relevant modules were detected. In addition, according to degree ≥ 10, 14 core genes including FOXM1, CXCR4, KPNA2, NANOG, UBE2C, KIF11, ZWINT, CDCA5, DLGAP5, KIF15, MCM2, MELK, SPP1, and TRIP13 were identified. Kaplan-Meier analysis, Oncomine, and Gene Expression Profiling Interactive Analysis showed that overexpression of FOXM1, SPP1, UBE2C, KIF11, ZWINT, CDCA5, UBE2C, and KIF15 was related to bad prognosis of EOC patients. CDCA5, FOXM1, KIF15, MCM2, and ZWINT were associated with stage. Receiver operating characteristic (ROC) curve showed that messenger RNA levels of these five genes exhibited better diagnostic efficiency for normal and tumor tissues. The Human Protein Atlas database was performed. The protein levels of these five genes were significantly higher in tumor tissues compared with normal tissues. Functional enrichment analysis suggested that all the hub genes played crucial roles in citrate cycle tricarboxylic acid cycle. Furthermore, the univariate and multivariate Cox proportional hazards regression showed that ZWINT was independent prognostic indictor among EOC patients. The genes and pathways discovered in the above studies may open a new direction for EOC treatment.
Collapse
Affiliation(s)
- Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huangyang Meng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Siyue Li
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yujie Shen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Wang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wu Shan
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiangnan Qiu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Zhang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Xing Z, Luo Z, Yang H, Huang Z, Liang X. Screening and identification of key biomarkers in adrenocortical carcinoma based on bioinformatics analysis. Oncol Lett 2019; 18:4667-4676. [PMID: 31611976 PMCID: PMC6781718 DOI: 10.3892/ol.2019.10817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 08/08/2019] [Indexed: 12/11/2022] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis. The presently available understanding of the pathogenesis of ACC is incomplete and the treatment options for patients with ACC are limited. Gene marker identification is required for accurate and timely diagnosis of the disease. In order to identify novel candidate genes associated with the occurrence and progression of ACC, the microarray datasets, GSE12368 and GSE19750, were obtained from Gene Expression Omnibus. Differentially expressed genes (DEGs) were identified, and functional enrichment analysis was performed. A protein-protein interaction network (PPI) was constructed to identify significantly altered modules, and module analysis was performed using Search Tool for the Retrieval of Interacting Genes and Cytoscape. A total of 228 DEGs were screened, consisting of 29 up and 199 downregulated genes. The enriched functions and pathways of the DEGs primarily included 'cell division', 'regulation of transcription involved in G1/S transition of mitotic cell cycle', 'G1/S transition of mitotic cell cycle', 'p53 signaling pathway' and 'oocyte meiosis'. A total of 14 hub genes were identified, and biological process analysis revealed that these genes were significantly enriched in cell division and mitotic cell cycle. Furthermore, survival analysis revealed that AURKA, TYMS, GINS1, RACGAP1, RRM2, EZH2, ZWINT, CDK1, CCNB1, NCAPG and TPX2 may be involved in the tumorigenesis, progression or prognosis of ACC. In conclusion, the 14 hub genes identified in the present study may aid researchers in elucidating the molecular mechanisms associated with the tumorigenesis and progression of ACC, and may be powerful and promising candidate biomarkers for the diagnosis and treatment of ACC.
Collapse
Affiliation(s)
- Zengmiao Xing
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zuojie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Haiyan Yang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhenxing Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xinghuan Liang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
24
|
Zw10 is a spindle assembly checkpoint protein that regulates meiotic maturation in mouse oocytes. Histochem Cell Biol 2019; 152:207-215. [DOI: 10.1007/s00418-019-01800-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 01/17/2023]
|
25
|
Integrative omics analysis identifies macrophage migration inhibitory factor signaling pathways underlying human hepatic fibrogenesis and fibrosis. JOURNAL OF BIO-X RESEARCH 2019; 2:16-24. [PMID: 32953199 PMCID: PMC7500331 DOI: 10.1097/jbr.0000000000000026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The genetic basis underlying liver fibrosis remains largely unknown. We conducted a study to identify genetic alleles and underlying pathways associated with hepatic fibrogenesis and fibrosis at the genome-wide level in 121 human livers. By accepting a liberal significance level of P<1e-4, we identified 73 and 71 candidate loci respectively affecting the variability in alpha-smooth muscle actin (α-SMA) levels (fibrogenesis) and total collagen content (fibrosis). The top genetic loci associated with the two markers were BAZA1 and NOL10 for α-SMA expression and FAM46A for total collagen content (P<1e-6). We further investigated the relationship between the candidate loci and the nearby gene transcription levels (cis-expression quantitative trait loci) in the same liver samples. We found that 44 candidate loci for α-SMA expression and 44 for total collagen content were also associated with the transcription of the nearby genes (P<0.05). Pathway analyses of these genes indicated that macrophage migration inhibitory factor (MIF) related pathway is significantly associated with fibrogenesis and fibrosis, though different genes were enriched for each marker. The association between the single nucleotide polymorphisms, MIF and α-SMA showed that decreased MIF expression is correlated with increased α-SMA expression, suggesting that variations in MIF locus might affect the susceptibility of fibrogenesis through controlling MIF gene expression. In summary, our study identified candidate alleles and pathways underlying both fibrogenesis and fibrosis in human livers. Our bioinformatics analyses suggested MIF pathway as a strong candidate involved in liver fibrosis, thus further investigation for the role of the MIF pathway in liver fibrosis is warranted. The study was reviewed and approved by the Institutional Review Board (IRB) of Wayne State University (approval No. 201842) on May 17, 2018.
Collapse
|
26
|
Peng F, Li Q, Niu SQ, Shen GP, Luo Y, Chen M, Bao Y. ZWINT is the next potential target for lung cancer therapy. J Cancer Res Clin Oncol 2019; 145:661-673. [PMID: 30643969 DOI: 10.1007/s00432-018-2823-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 12/12/2018] [Indexed: 11/30/2022]
Abstract
PURPOSE We aimed to analyze the expression of ZWINT, NUSAP1, DLGAP5, and PRC1 in tumor tissues and adjacent tissues with public data. METHODS The expression patterns of four genes were detected in cancer tissues and adjacent tissues by qRT-PCR. The overall survival analysis was used to explore these genes in lung adenocarcinoma and squamous cell carcinoma patients. Knockdown assays were used to select the most suitable gene among these four genes. Cell function assays with the knockdown gene were conducted in A549 and NCL H226 cells. The role of the knockdown gene in lung cancer was dissected in a mice tumor model. Transcriptome sequencing analyses with the knockdown gene were analyzed. RESULTS Overexpression of these genes was significantly detected in cancer tissues (P < 0.01). Overall survival revealed that high expression of these genes is closely related with poor prognosis of lung adenocarcinoma patients (P < 0.05). Knockdown of ZWINT reduced proliferation in NCI H226 and A549 cells (P < 0.05). Knockdown also inhibited cell migration, invasion, apoptosis, and colony formation (P < 0.05). ZWINT knockdown reduced tumor volume (P < 0.05). Transcriptome sequencing of ZWINT knockdown-treated A549 and NCI H226 cells indicated that 100 and 426 differentially expressed genes were obtained, respectively. Gene ontology analysis suggested that binding, biological regulation, and multicellular organismal processes were the most enriched. KEGG analysis revealed that TNF, P53, and PI3K signal networks would be the most potential ZWINT-related pathways and were identified by Western blot analysis. CONCLUSIONS ZWINT may be a novel target for lung cancer therapy.
Collapse
Affiliation(s)
- Fang Peng
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Qiang Li
- Department of Organ Transplantation and General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Shao-Qing Niu
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Guo-Ping Shen
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Ying Luo
- Department of Clinical Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Ming Chen
- Department of Radiation Oncology, Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, 1 East Banshan Road, Hangzhou, 310022, Zhejiang, People's Republic of China.
| | - Yong Bao
- Department of Radiation Oncology, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
27
|
Suman S, Mishra A. An interaction network driven approach for identifying biomarkers for progressing cervical intraepithelial neoplasia. Sci Rep 2018; 8:12927. [PMID: 30150654 PMCID: PMC6110773 DOI: 10.1038/s41598-018-31187-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 08/10/2018] [Indexed: 12/13/2022] Open
Abstract
Overlapping genes across high-grade squamous intraepithelial lesions (CIN2 and 3) and cancer may serve as potential biomarkers for this progressive disease. Differentially expressed genes (DEGs) of dysplastic (CIN2 and CIN3) and cancer cells were identified by microarray data analysis. Gene interaction network was constructed using the 98 common DEGs among the dysplastic and cancer cells and analysed for the identification of common modules, hubs and significant motifs. Two significant modules and 10 hubs of the common gene interaction network, with 125 nodes and 201 edges were found. DEGs namely NDC80, ZWINT, CDC7, MCM4, MCM2 and MCM6 were found to be common in both the significant modules as well as the hubs. Of these, ZWINT, CDC7, MCM4, MCM2 and MCM6 were further identified to be part of most significant motifs. This overlapping relationship provides a list of common disease related genes among pre-cancerous and cancer stages which could help in targeting the proliferating cancerous cells during onset. Capitalizing upon and targeting Minichromosome maintenance protein complex - specifically the MCM2, MCM4 and MCM6 subunits, ZWINT and CDC7 for experimental validation, may provide valuable insights in understanding and detection of progressing cervical neoplasia to cervical cancer at an early stage.
Collapse
Affiliation(s)
- Shikha Suman
- Division of Applied Sciences, Indian Institute of Information Technology (IIIT), Allahabad, 211012, India.
| | - Ashutosh Mishra
- Division of Applied Sciences, Indian Institute of Information Technology (IIIT), Allahabad, 211012, India
| |
Collapse
|
28
|
Alaraudanjoki VK, Koivisto S, Pesonen P, Männikkö M, Leinonen J, Tjäderhane L, Laitala ML, Lussi A, Anttonen VAM. Genome-Wide Association Study of Erosive Tooth Wear in a Finnish Cohort. Caries Res 2018; 53:49-59. [PMID: 29898447 DOI: 10.1159/000488208] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/25/2018] [Indexed: 11/19/2022] Open
Abstract
Erosive tooth wear is defined as irreversible loss of dental tissues due to intrinsic or extrinsic acids, exacerbated by mechanical forces. Recent studies have suggested a higher prevalence of erosive tooth wear in males, as well as a genetic contribution to susceptibility to erosive tooth wear. Our aim was to examine erosive tooth wear by performing a genome-wide association study (GWAS) in a sample of the Northern Finland Birth Cohort 1966 (n = 1,962). Erosive tooth wear was assessed clinically using the basic erosive wear examination. A GWAS was performed for the whole sample as well as separately for males and females. We identified one genome-wide significant signal (rs11681214) in the GWAS of the whole sample near the genes PXDN and MYT1L. When the sample was stratified by sex, the strongest genome-wide significant signals were observed in or near the genes FGFR1, C8orf86, CDH4, SCD5, F2R, and ING1. Additionally, multiple suggestive association signals were detected in all GWASs performed. Many of the signals were in or near the genes putatively related to oral environment or tooth development, and some were near the regions considered to be associated with dental caries, such as 2p24, 4q21, and 13q33. Replications of these associations in other samples, as well as experimental studies to determine the biological functions of associated genetic variants, are needed.
Collapse
Affiliation(s)
| | - Salla Koivisto
- Research Unit of Oral Health Sciences, University of Oulu, Oulu, Finland
| | - Paula Pesonen
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Minna Männikkö
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Jukka Leinonen
- Research Unit of Oral Health Sciences, University of Oulu, Oulu, Finland
| | - Leo Tjäderhane
- Research Unit of Oral Health Sciences, University of Oulu, Oulu, Finland.,Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Adrian Lussi
- Department of Preventive, Restorative and Pediatric Dentistry, University of Bern, Bern, Switzerland
| | | |
Collapse
|
29
|
Huang R, Gao L. Identification of potential diagnostic and prognostic biomarkers in non-small cell lung cancer based on microarray data. Oncol Lett 2018; 15:6436-6442. [PMID: 29731852 PMCID: PMC5921217 DOI: 10.3892/ol.2018.8153] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most commonly diagnosed subtype of lung cancer, and the leading cause of cancer-associated mortalities worldwide. However, NSCLC is typically diagnosed at a late stage of disease due to a lack of effective diagnostic methods. In the present study, the GSE19804 dataset was obtained from the Gene Expression Omnibus, and a number of differentially expressed genes were identified between NSCLC and adjacent normal tissues. Based on functional and pathway enrichment analyses, five hub genes (cell-division cycle 20, centromere protein F, kinesin family member 2C, BUB1 mitotic checkpoint serine/threonine kinase and ZW10 interacting kinetochore protein) were selected. After verifying that the mRNA level of these hub genes was also upregulated in NSCLC tissues by using the GSE10072 dataset and in cell lines by reverse transcription-quantitative polymerase chain reaction. The diagnostic and prognostic potentials of these five gene candidates were evaluated using receiver operating characteristic curves and survival analyses. Taken together, the present study identified five candidates that are overexpressed in NSCLC tissues and could also serve as potential diagnostic and prognostic biomarkers for patients with NSCLC.
Collapse
Affiliation(s)
- Ru Huang
- Department of Heart Failure, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Lei Gao
- Department of Heart Failure, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China.,Key Laboratory of Arrhythmias of The Ministry of Education of China, Research Institute of Heart Failure, Shanghai East Hospital, Dalian Medical University, Shanghai 200120, P.R. China
| |
Collapse
|
30
|
Spc24 is required for meiotic kinetochore-microtubule attachment and production of euploid eggs. Oncotarget 2018; 7:71987-71997. [PMID: 27713128 PMCID: PMC5342138 DOI: 10.18632/oncotarget.12453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/29/2016] [Indexed: 01/29/2023] Open
Abstract
Mammalian oocytes are particularly error prone in chromosome segregation during two successive meiotic divisions. The proper kinetochore-microtubule attachment is a prerequisite for faithful chromosome segregation during meiosis. Here, we report that Spc24 localizes at the kinetochores during mouse oocyte meiosis. Depletion of Spc24 using specific siRNA injection caused defective kinetochore-microtubule attachments and chromosome misalignment, and accelerated the first meiosis by abrogating the kinetochore recruitment of spindle assembly checkpoint protein Mad2, leading to a high incidence of aneuploidy. Thus, Spc24 plays an important role in genomic stability maintenance during oocyte meiotic maturation.
Collapse
|
31
|
Ying H, Xu Z, Chen M, Zhou S, Liang X, Cai X. Overexpression of Zwint predicts poor prognosis and promotes the proliferation of hepatocellular carcinoma by regulating cell-cycle-related proteins. Onco Targets Ther 2018; 11:689-702. [PMID: 29440916 PMCID: PMC5800459 DOI: 10.2147/ott.s152138] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Zwint, a centromere-complex component required for the mitotic spindle checkpoint, has been reported to be overexpressed in different human cancers, but it has not been studied in human hepatocellular carcinoma (HCC). Materials and methods The role of Zwint in hepatocellular carcinoma cell proliferation capacities was evaluated by using cell counting kit-8 (CCK8), flow cytometry, clone formation and tumor formation assay in nude mice. Western blot analysis and qPCR assay were performed to assess Zwint interacting with cell-cycle-related proteins. Results We report that ZWINT mRNA and protein expression were upregulated in HCC samples and cell lines. An independent set of 106 HCC-tissue pairs and corresponding noncancerous tissues was evaluated for Zwint expression using immunohistochemistry, and elevated Zwint expression in HCC tissues was significantly correlated with clinicopathological features, such as tumor size and number. Kaplan–Meier survival and Cox regression analysis revealed that high expression of Zwint was correlated with poor overall survival and a greater tendency for tumor recurrence. Ectopic expression of Zwint promoted HCC-cell proliferation, and Zwint expression affected the expression of several cell-cycle proteins, including PCNA, cyclin B1, Cdc25C and CDK1. Conclusion Our findings suggest that upregulation of Zwint may contribute to the progression of HCC and may be a prognostic biomarker and potential therapeutic target for treating HCC.
Collapse
Affiliation(s)
- Hanning Ying
- Department of General Surgery.,Key Laboratory of Endoscopic Technique Research of Zhejiang Province
| | - Zhiyao Xu
- Central Lab of Biomedical Research Center, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Mingming Chen
- Department of General Surgery.,Key Laboratory of Endoscopic Technique Research of Zhejiang Province
| | - Senjun Zhou
- Department of General Surgery.,Key Laboratory of Endoscopic Technique Research of Zhejiang Province
| | - Xiao Liang
- Department of General Surgery.,Key Laboratory of Endoscopic Technique Research of Zhejiang Province
| | - Xiujun Cai
- Department of General Surgery.,Key Laboratory of Endoscopic Technique Research of Zhejiang Province
| |
Collapse
|
32
|
Wang HJ, Wang L, Lv J, Fu LQ, Wang Z, He XL, Ma YY, Li L, Zhao TW, Xu XG, Yu LL, Pan HY, Hu ZM, Mou XZ. Decreased expression of Zwint-1 is associated with poor prognosis in hepatocellular carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10406-10412. [PMID: 31966377 PMCID: PMC6965758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/25/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND AIM ZW10 interacting kinetochore protein 1 (Zwint-1), one of the major kinetochore proteins, is essential for kinetochore function, such as spindle assembly checkpoint function and kinetochore-microtubule attachment. Recently, it has been found over-expressed in some human cancers, including ovarian cancer, bladder cancer, and pulmonary adenocarcinoma. However, few studies of the expression of Zwint-1 in hepatocellular carcinoma (HCC) have been reported. This study is aimed to investigate the expression of Zwint-1 and its relationship with clinical pathological characters in HCC. METHODS The expression of Zwint-1 protein was analyzed by immunohistochemistry staining on tissue microarrays containing 171 HCC tissues and 187 control non-tumorous liver tissues. The relationships between the Zwint-1 expression and the clinicopathological parameters, and survival analysis were investigated using SPSS software 13.0. RESULTS Zwint-1 was found uniformly expressed in adjacent non-tumorous liver tissues (184/187, 98.40%), while was significantly decreased in HCC tissues, or even absent (150 of 171, 61.82%, P<0.001). The expression of Zwint-1 was negatively associated with age, tumor size, and Edmondson Grade. Besides, HCC patients with low Zwint-1 expression were also correlated with poor overall survival of the patients. CONCLUSIONS Decreased expression of Zwint-1 was associated with poor prognosis in HCC.
Collapse
Affiliation(s)
- Hui-Ju Wang
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang ProvinceHangzhou, Zhejiang, China
| | - Liang Wang
- Department of Hand Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Jia Lv
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Luo-Qin Fu
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Zhen Wang
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Xiang-Lei He
- Department of Pathology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Ying-Yu Ma
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang ProvinceHangzhou, Zhejiang, China
| | - Li Li
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
- Key Laboratory of Gastroenterology of Zhejiang ProvinceHangzhou, Zhejiang, China
| | - Tong-Wei Zhao
- Department of Medical Oncology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Xiao-Gang Xu
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang UniversityHangzhou, China
| | - Li-Li Yu
- Department of Pathology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Hong-Ying Pan
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Zhi-Ming Hu
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical CollegeHangzhou, Zhejiang, China
- School of Basic Medical Sciences, Hangzhou Medical CollegeHangzhou, Zhejiang, China
| |
Collapse
|
33
|
Agarwal R, Narayan J, Bhattacharyya A, Saraswat M, Tomar AK. Gene expression profiling, pathway analysis and subtype classification reveal molecular heterogeneity in hepatocellular carcinoma and suggest subtype specific therapeutic targets. Cancer Genet 2017; 216-217:37-51. [PMID: 29025594 DOI: 10.1016/j.cancergen.2017.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/12/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023]
Abstract
A very low 5-year survival rate among hepatocellular carcinoma (HCC) patients is mainly due to lack of early stage diagnosis, distant metastasis and high risk of postoperative recurrence. Hence ascertaining novel biomarkers for early diagnosis and patient specific therapeutics is crucial and urgent. Here, we have performed a comprehensive analysis of the expression data of 423 HCC patients (373 tumors and 50 controls) downloaded from The Cancer Genome Atlas (TCGA) followed by pathway enrichment by gene ontology annotations, subtype classification and overall survival analysis. The differential gene expression analysis using non-parametric Wilcoxon test revealed a total of 479 up-regulated and 91 down-regulated genes in HCC compared to controls. The list of top differentially expressed genes mainly consists of tumor/cancer associated genes, such as AFP, THBS4, LCN2, GPC3, NUF2, etc. The genes over-expressed in HCC were mainly associated with cell cycle pathways. In total, 59 kinases associated genes were found over-expressed in HCC, including TTK, MELK, BUB1, NEK2, BUB1B, AURKB, PLK1, CDK1, PKMYT1, PBK, etc. Overall four distinct HCC subtypes were predicted using consensus clustering method. Each subtype was unique in terms of gene expression, pathway enrichment and median survival. Conclusively, this study has exposed a number of interesting genes which can be exploited in future as potential markers of HCC, diagnostic as well as prognostic and subtype classification may guide for improved and specific therapy.
Collapse
Affiliation(s)
- Rahul Agarwal
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Jitendra Narayan
- Unité de recherche en biologie environnementale et évolutive (URBE), University of Namur, Belgium
| | | | - Mayank Saraswat
- Transplantation Laboratory, Haartmaninkatu 3, University of Helsinki, Helsinki, Finland
| | - Anil Kumar Tomar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
34
|
Xu Z, Zhou Y, Cao Y, Dinh TLA, Wan J, Zhao M. Identification of candidate biomarkers and analysis of prognostic values in ovarian cancer by integrated bioinformatics analysis. Med Oncol 2016; 33:130. [PMID: 27757782 DOI: 10.1007/s12032-016-0840-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/05/2016] [Indexed: 12/28/2022]
Abstract
Ovarian cancer is the first leading cause of mortality in gynecological malignancies. To identify key genes and microRNAs in ovarian cancer, mRNA microarray dataset GSE36668, GSE18520, GSE14407 and microRNA dataset GSE47841 were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) and microRNAs (DEMs) were obtained using GEO2R. Functional and pathway enrichment analysis were performed for DEGs using DAVID database. Protein-protein interaction (PPI) network was established by STRING and visualized by Cytoscape. Following, overall survival (OS) analysis of hub genes was performed by the Kaplan-Meier plotter online tool. Module analysis of the PPI network was performed using MCODE. Moreover, miRecords was applied to predict the targets of the DEMs. A total of 345 DEGs were obtained, which were mainly enriched in the terms related to cell cycle, mitosis, and ovulation cycle process. A PPI network was constructed, consisting of 141 nodes and 296 edges. Sixteen genes had high degrees in the network. High expression of four genes of the 16 genes was associated with worse OS of patients with ovarian cancer, including CCNB1, CENPF, KIF11, and ZWINT. A significant module was detected from the PPI network. The enriched functions and pathways included cell cycle, nuclear division, and oocyte meiosis. Additionally, a total of 36 DEMs were identified. The expression of KIF11 was negatively correlated with that of has-miR-424 and has-miR-381, and it was also the potential target of two microRNAs. In conclusion, these results identified key genes, which could provide potential targets for ovarian cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Zhanzhan Xu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China
| | - Yu Zhou
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China
| | - Yexuan Cao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Thi Lan Anh Dinh
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China
| | - Min Zhao
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China.
| |
Collapse
|
35
|
Uematsu K, Okumura F, Tonogai S, Joo-Okumura A, Alemayehu DH, Nishikimi A, Fukui Y, Nakatsukasa K, Kamura T. ASB7 regulates spindle dynamics and genome integrity by targeting DDA3 for proteasomal degradation. J Cell Biol 2016; 215:95-106. [PMID: 27697924 PMCID: PMC5057283 DOI: 10.1083/jcb.201603062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/30/2016] [Indexed: 12/31/2022] Open
Abstract
Uematsu et al. show that ASB7 ubiquitinates DDA3, which facilitates Kif2a-mediated depolymerization of microtubules (MTs) for proteasomal degradation. The presence of MTs prevents the ASB7–DDA3 interaction, suggesting a feedback loop to appropriately regulate MT polymerization and spindle dynamics. Proper dynamic regulation of the spindle is essential for successful cell division. However, the molecular mechanisms that regulate spindle dynamics in mitosis are not fully understood. In this study, we show that Cullin 5–interacting suppressor of cytokine signaling box protein ASB7 ubiquitinates DDA3, a regulator of spindle dynamics, thereby targeting it for proteasomal degradation. The presence of microtubules (MTs) prevented the ASB7–DDA3 interaction, thus stabilizing DDA3. Knockdown of ASB7 decreased MT polymerization and increased the proportion of cells with unaligned chromosomes, and this phenotype was rescued by deletion of DDA3. Collectively, these data indicate that ASB7 plays a crucial role in regulating spindle dynamics and genome integrity by controlling the expression of DDA3.
Collapse
Affiliation(s)
- Keiji Uematsu
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Fumihiko Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Syunsuke Tonogai
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Akiko Joo-Okumura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Dawit Hailu Alemayehu
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Akihiko Nishikimi
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan Research Center for Advanced Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunio Nakatsukasa
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| | - Takumi Kamura
- Division of Biological Science, Graduate School of Science, Nagoya University, Aichi 464-8602, Japan
| |
Collapse
|
36
|
Yuan YF, Ren YX, Yuan P, Yan LY, Qiao J. TRAIP is involved in chromosome alignment and SAC regulation in mouse oocyte meiosis. Sci Rep 2016; 6:29735. [PMID: 27405720 PMCID: PMC4942609 DOI: 10.1038/srep29735] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/22/2016] [Indexed: 12/14/2022] Open
Abstract
Recent whole-exome sequencing (WES) studies demonstrated that TRAIP is associated with primordial dwarfism. Although TRAIP was partially studied in mitosis, its function in oocyte meiosis remained unknown. In this study, we investigated the roles of TRAIP during mouse oocyte meiosis. TRAIP was stably expressed during oocytes meiosis and co-localized with CREST at the centromere region. Knockdown of TRAIP led to DNA damage, as revealed by the appearance of γH2AX. Although oocytes meiotic maturation was not affected, the proportions of misaligned chromosomes and aneuploidy were elevated after TRAIP knockdown, suggesting TRAIP is required for stable kinetochore–microtubule (K-MT) attachment. TRAIP knockdown decreased the accumulation of Mad2 on centromeres, potentially explaining why oocyte maturation was not affected following formation of DNA lesions. Securin, a protein which was prevent from precocious degradation by Mad2, was down-regulated after TRAIP knockdown. Inhibition of TRAIP by microinjection of antibody into pro-metaphase I (pro-MI) stage oocytes resulted in precocious first polar body (PB1) extrusion, and live-cell imaging clearly revealed misaligned chromosomes after TRAIP knockdown. Taken together, these data indicate that TRAIP plays important roles in oocyte meiosis regulation.
Collapse
Affiliation(s)
- Yi-Feng Yuan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, HaiDian District, Beijing 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| | - Yi-Xin Ren
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| | - Peng Yuan
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| | - Li-Ying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, HaiDian District, Beijing 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, No. 49 North HuaYuan Road, HaiDian District, Beijing 100191, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| |
Collapse
|
37
|
You SY, Park YS, Jeon HJ, Cho DH, Jeon HB, Kim SH, Chang JW, Kim JS, Oh JS. Beclin-1 knockdown shows abscission failure but not autophagy defect during oocyte meiotic maturation. Cell Cycle 2016; 15:1611-9. [PMID: 27149384 DOI: 10.1080/15384101.2016.1181235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cytokinesis is the final step in cell division that results in the separation of a parent cell into daughter cells. Unlike somatic cells that undergo symmetric division, meiotic division is highly asymmetric, allowing the preservation of maternal resources for embryo development. Beclin-1/BECN1, the mammalian homolog of yeast Atg6, is a key molecule of autophagy. As part of a class III phosphatidylinositol 3-kinase (PI3K-III) complex, BECN1 initiates autophagosome formation by coordinating membrane trafficking. However, emerging evidence suggests that BECN1 regulates chromosome segregation and cytokinesis during mitosis. Thus, we investigated the function of BECN1 during oocyte meiotic maturation. BECN1 was widely distributed during meiotic maturation forming small vesicles. Interestingly, BECN1 is also detected at the midbody ring during cytokinesis. Depletion of BECN1 impaired the cytokinetic abscission, perturbing the recruitment of ZFYVE26 at the midbody. Similar phenotypes were observed when PI3K-III activity was inhibited. However, inhibition of autophagy by depleting Atg14L did not disturb meiotic maturation. Therefore, our results not only demonstrate that BECN1 as a PI3K-III component is essential for cytokinesis, but also suggest that BECN1 is not associated with autophagy pathway in mouse oocytes.
Collapse
Affiliation(s)
- Seung Yeop You
- a Department of Genetic Engineering , College of Biotechnology and Bioengineering, Sungkyunkwan University , Suwon , Gyeonggi-do , Korea
| | - Yong Seok Park
- a Department of Genetic Engineering , College of Biotechnology and Bioengineering, Sungkyunkwan University , Suwon , Gyeonggi-do , Korea
| | - Hyuk-Joon Jeon
- a Department of Genetic Engineering , College of Biotechnology and Bioengineering, Sungkyunkwan University , Suwon , Gyeonggi-do , Korea
| | - Dong-Hyung Cho
- b Department of East-West Medical Science , Graduate School of East-West Medical Science, Kyung Hee University , Yongin , Korea
| | - Hong Bae Jeon
- c Biomedical Research Institute, MEDIPOST Co., Ltd. , Seongnam , Korea
| | - Sung Hyun Kim
- d Department of Neuroscience , Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University , Seoul , Korea
| | - Jong Wook Chang
- e Department of Health Sciences and Technology , Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University , Seoul , Korea
| | - Jae-Sung Kim
- f Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences , Seoul , Korea
| | - Jeong Su Oh
- a Department of Genetic Engineering , College of Biotechnology and Bioengineering, Sungkyunkwan University , Suwon , Gyeonggi-do , Korea
| |
Collapse
|