1
|
Wang L, He X, Shen Y, Chen J, Chen Y, Zhou Z, Xu X. A Novel Ferroptosis-Related Gene Prognosis Signature and Identifying Atorvastatin as a Potential Therapeutic Agent for Hepatocellular Carcinoma. Curr Issues Mol Biol 2025; 47:201. [PMID: 40136455 PMCID: PMC11940908 DOI: 10.3390/cimb47030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Among the most common malignant tumors, hepatocellular carcinoma (HCC) is a primary liver cancer type that has a high mortality rate. HCC often presents insidiously, is prone to recurrence, and has limited treatment efficacy. Ferroptosis regulates tumorigenesis, progression, and metastasis, which is a novel form of iron-dependent cell death. Numerous studies suggest that HCC is sensitive to ferroptosis, indicating that targeted therapies aimed at inducing ferroptosis may represent a promising new approach to cancer treatment. This study aims to find genes associated with HCC and ferroptosis, as well as to screen for potential agents that may cause ferroptosis in HCC. Transcriptome and clinical sample data were obtained from the TCGA database to identify differentially expressed genes related to ferroptosis. Using various regression and survival analysis techniques, we developed a prognostic model based on four core genes and evaluated its predictive potential. Subsequently, we screened for potential therapeutic agents in the Connective Map (CMap) database, designated as compound Atorvastatin, based on differential genes from two risk groups and related to ferroptosis. Through experiments conducted in vivo and in vitro, we demonstrated that Atorvastatin can induce ferroptosis in HCC cells while inhibiting their growth and migration. In conclusion, this research targets ferroptosis therapy and provides new insights for improving the prediction and prevention of HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.W.); (X.H.); (Y.S.); (J.C.); (Y.C.); (Z.Z.)
| |
Collapse
|
2
|
Li J, Liu L, Zong G, Yang Z, Zhang D, Zhao B. Knockdown of CENPF induces cell cycle arrest and inhibits epithelial‑mesenchymal transition progression in glioma. Oncol Lett 2025; 29:61. [PMID: 39611064 PMCID: PMC11602827 DOI: 10.3892/ol.2024.14807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/02/2024] [Indexed: 11/30/2024] Open
Abstract
Gliomas are among the most common malignant tumors of the central nervous system. Despite surgical resection followed by postoperative radiotherapy and chemotherapy, their prognosis remains unfavorable. The present study aimed to assess new mechanisms and explore promising prognostic biomarkers for patients with glioma using comprehensive bioinformatics analysis and in vitro and in vivo assays. Overlapping differentially expressed genes were screened from The Cancer Genome Atlas, GSE111260 and GSE16011 samples for protein-protein interaction networks, a risk score model, gene mutation analysis and a nomogram to identify the prognostic hub genes. Subsequently, an immunoassay was performed to determine key genes. Functional and animal assays were then performed to assess the tumorigenesis of the key genes in glioma. Using bioinformatics analysis, centromere protein F (CENPF), kinesin superfamily member 20A, kinesin superfamily protein 4A and marker of proliferation Ki-67 were identified as potential prognostic biomarkers for patients with glioma. Furthermore, CENPF knockdown was demonstrated to suppress the proliferation and metastasis of glioma cells, and induce G2 arrest in the cell cycle. Moreover, CENPF knockdown was revealed to decrease Vimentin and increase E-cadherin levels in glioma cells, and significantly reduce the size and mass of tumors in vivo. Overall, the present study identified new clinical biomarkers and revealed that CENPF may promote glioma progression by regulating the epithelial-mesenchymal transition pathway. By elucidating the complexities of glioma and identifying prognostic biomarkers, the present research enables further improvement of patient outcomes and the advancement of precision medicine for this disease.
Collapse
Affiliation(s)
- Jia Li
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Lei Liu
- Department of Emergency Surgery, Fuyang Hospital Affiliated to Anhui Medical University, Fuyang, Anhui 236112, P.R. China
| | - Gang Zong
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Deran Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
3
|
Li Y, Ge S, Liu J, Sun D, Xi Y, Chen P. Nuclear Structure, Size Regulation, and Role in Cell Migration. Cells 2024; 13:2130. [PMID: 39768219 PMCID: PMC11675058 DOI: 10.3390/cells13242130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
The nucleus serves as a pivotal regulatory and control hub in the cell, governing numerous aspects of cellular functions, including DNA replication, transcription, and RNA processing. Therefore, any deviations in nuclear morphology, structure, or organization can strongly affect cellular activities. In this review, we provide an updated perspective on the structure and function of nuclear components, focusing on the linker of nucleoskeleton and cytoskeleton complex, the nuclear envelope, the nuclear lamina, and chromatin. Additionally, nuclear size should be considered a fundamental parameter for the cellular state. Its regulation is tightly linked to environmental changes, development, and various diseases, including cancer. Hence, we also provide a concise overview of different mechanisms by which nuclear size is determined, the emerging role of the nucleus as a mechanical sensor, and the implications of altered nuclear morphology on the physiology of diseased cells.
Collapse
Affiliation(s)
- Yuhao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China;
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China (D.S.); (Y.X.)
| | - Shanghao Ge
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China (D.S.); (Y.X.)
| | - Jiayi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China (D.S.); (Y.X.)
| | - Deseng Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China (D.S.); (Y.X.)
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China (D.S.); (Y.X.)
| | - Pan Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China;
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo 315211, China (D.S.); (Y.X.)
| |
Collapse
|
4
|
Su Z, Zhong Y, He Y, You L, Xin F, Wang L, Liu Z. Bulk- and single cell-RNA sequencing reveal KIF20A as a key driver of hepatocellular carcinoma progression and immune evasion. Front Immunol 2024; 15:1469827. [PMID: 39555078 PMCID: PMC11563802 DOI: 10.3389/fimmu.2024.1469827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Kinesin family member 20A (KIF20A) is essential for cell proliferation and is implicated in promoting tumor progression, but its role in hepatocellular carcinoma (HCC) remains poorly studied. Methods Through the analysis of bulk RNA-sequencing (bulk RNA-seq) and single-cell RNA sequencing (scRNA-seq) data, the expression of KIF20A and its relationship with diagnosis, prognosis, and the immune microenvironment were examined. The association between KIF20A and the malignant progression and metastasis of HCC was confirmed through in vitro and in vivo experiments. Furthermore, patient re-staging was performed using Recursive Partitioning Analysis (RPA) to enhance clinical benefit. Results In this study, we firstly found KIF20A was overexprerssed in HCC both by bulk RNA-seq and scRNA-seq, and then the overexpression of KIF20A significantly promoted the proliferation, invasion, and metastasis in vitro. In vivo, the overexpression of KIF20A promoted the growth and lung metastasis of HCC. Furthermore, gene set variation analysis of bulk RNA-seq and scRNA-seq revealed that KIF20A might be associated with cell cycle related signaling pathways of E2F and G2M, and overexpression of KIF20A inhibited the activity of p21 and bax, as well as shortened G2 phase. Importantly, we found that KIF20A could induce T cell exhaustion via the SPP1-CD44 axe using scRNA-seq. Additionally, KIF20A was also correlated with the expression of immune checkpoint inhibitors (ICIs), and KIF20Ahigh subgroup might be benefited from the ICIs therapy. Conclusion KIF20A emerges as a pivotal driver of HCC progression, intricately regulating cell cycle pathways and modulating immune responses, which position KIF20A as a promising target for HCC management.
Collapse
Affiliation(s)
- Zhixiong Su
- Department of Radiation Oncology, Jiangxi Clinical Research Center for Cancer, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yaqi Zhong
- Department of Hepatopancreatobiliary Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yufang He
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Lijie You
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Fuli Xin
- Department of Hepatopancreatobiliary Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Lei Wang
- Department of Radiation Oncology, Jiangxi Clinical Research Center for Cancer, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Zhihua Liu
- Department of Radiation Oncology, Jiangxi Clinical Research Center for Cancer, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Ghnim ZS, Mahdi MS, Ballal S, Chahar M, Verma R, Al-Nuaimi AMA, Kumar MR, Al-Hussein RKA, Adil M, Jawad MJ. The role of kinesin superfamily proteins in hepatocellular carcinoma. Med Oncol 2024; 41:271. [PMID: 39400594 DOI: 10.1007/s12032-024-02497-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024]
Abstract
The most prevalent form of primary liver cancer, hepatocellular carcinoma (HCC) poses a significant global health challenge due to its limited therapeutic options. Researchers are currently focused on the complex molecular landscape that governs the initiation and progression of HCC in order to identify new avenues for diagnosis, prognosis, and treatment. In the context of HCC, the Kinesin Superfamily Proteins (KIFs) have become critical regulators of cellular processes, prompting a growing interest in their function among the diverse array of molecular actors implicated in cancer. The KIFs, a family of microtubule-based molecular motors, are renowned for their essential roles in the dynamics of mitotic spindles and intracellular transport. Beyond their well-established functions in normal cellular physiology, emerging evidence indicates that dysregulation of KIFs significantly contributes to the pathogenesis of HCC. Novel therapeutic targets and diagnostic markers are revealed through the unique opportunity to comprehend the complex interplay between KIFs and the molecular events that drive HCC.
Collapse
Affiliation(s)
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India.
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University, Jaipur, Rajasthan, India
| | - Rajni Verma
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Amritsar, Punjab, 140307, India
| | | | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | | | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
6
|
Li J, Su P, Li T, Hao Y, Wang T, Fu L, Liu X. The Role and Clinical Relevance of Glycolysis-Associated Genes on Immune Infiltration in Hepatocellular Carcinoma. J Cell Biochem 2024; 125:e30620. [PMID: 38923014 DOI: 10.1002/jcb.30620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Hepatocellular carcinoma (HCC) poses a significant challenge with dismal survival rates, necessitating a deeper understanding of its molecular mechanisms and the development of improved therapies. Metabolic reprogramming, particularly heightened glycolysis, plays a crucial role in HCC progression. Glycolysis-associated genes (GAGs) emerge as key players in HCC pathogenesis, influencing the tumor microenvironment and immune responses. This study aims to investigate the intricate interplay between GAGs and the immune landscape within HCC, offering valuable insights into potential prognostic markers and therapeutic targets to enhance treatment strategies and patient outcomes. Through the exploration of GAGs, we have identified two distinct molecular glycolytic subtypes in HCC patients, each exhibiting significant differences in both the immune microenvironment and prognosis. A risk model comprising five key GAGs was formulated and subsequently evaluated for their predictive accuracy. Our findings underscore the diverse tumor microenvironment and immune responses associated with the varying glycolytic subtypes observed in HCC. The identified key GAGs hold promise as prognostic indicators for evaluating HCC risk levels, predicting patient outcomes, and guiding clinical treatment decisions, particularly in the context of anticipating responses to immunotherapy drugs.
Collapse
Affiliation(s)
- Jing Li
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Peng Su
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Ting Li
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yang Hao
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Tianjun Wang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Xin Liu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| |
Collapse
|
7
|
Yang J, Xu L, Han X. KIF20B Correlates with LUAD Progression and Is an Independent Risk Factor. Crit Rev Eukaryot Gene Expr 2024; 34:49-59. [PMID: 38305288 DOI: 10.1615/critreveukaryotgeneexpr.2023050271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Kinesin family proteins (KIFs) play crucial roles in human tumorigenesis and progression. This study aimed to investigate the expression and association of Kinesin family member 20B (KIF20B) with lung adenocarcinoma (LUAD). METHODS RNA-seq data from LUAD patients (n = 535) were extracted from TCGA. KIF20B expression was compared between tumor tissues and controls, and between different stages of the disease. Survival and Cox regression analyses were performed, as well as in vitro cellular experiments on A549 cells. RESULTS KIF20B is upregulated in LUAD tumor tissues compared with controls and is higher in advanced stages. Patients with high expression of KIF20B have shorter survival times. KIF20B is an independent risk factor for the prognosis of LUAD. High KIF20B expression samples were enriched in signaling pathways related to tumor progression. si-KIF20B transfection reduced migration and invasion of A549 cells and increased apoptosis. The expression of p53 and Bax proteins was upregulated by si-KIF20B, while Bcl-2 was down-regulated. DISCUSSION This study reveals that high KIF20B expression is an independent risk factor for the poor prognosis of LUAD. The inhibition of KIF20B might be of great value for suppressing LUAD progression.
Collapse
Affiliation(s)
- Jianye Yang
- Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital)
| | - Liang Xu
- Respiratory Medicine, Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital), No. 999, Zhongxing South Road, Shaoxing 312000, China
| | - Xiaoliang Han
- Affiliated Hospital of Shaoxing University (The Shaoxing Municipal Hospital)
| |
Collapse
|
8
|
Liu G, Zhang S, Lin R, Cao X, Yuan L. Anti-tumor target screening of sea cucumber saponin Frondoside A: a bioinformatics and molecular docking analysis. Front Oncol 2023; 13:1307838. [PMID: 38144520 PMCID: PMC10739435 DOI: 10.3389/fonc.2023.1307838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Cancer remains the leading cause of death worldwide. In spite of significant advances in targeted and immunotherapeutic approaches, clinical outcomes for cancer remain poor. The aim of the present study was to investigate the potential mechanisms and therapeutic targets of Frondoside A for the treatment of liver, pancreatic, and bladder cancers. The data presented in our study demonstrated that Frondoside A reduced the viability and migration of HepG2, Panc02, and UM-UC-3 cancer cell in vitro. Moreover, we utilized the GEO database to screen and identify for differentially expressed genes (DEGs) in liver, pancreatic, and bladder cancers, which resulted in the identification of 714, 357, and 101 DEGs, respectively. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation were performed using the Metascape database for DEGs that were significantly associated with cancer development. The protein-protein interaction (PPI) networks of the identified DEGs in liver, pancreatic, and bladder cancers were analyzed using Cytoscape 3.9.0 software, and subsequently identified potential key genes that were associated with these networks. Subsequently, their prognostic values were assessed by gene expression level analysis and Kaplan-Meier survival analysis (GEPIA). Furthermore, we utilized TIMER 2.0 to investigate the correlation between the expression of the identified key gene and cancer immune infiltration. Finally, molecular docking simulations were performed to assess the affinity of Frondoside A and key genes. Our results showed a significant correlation between these DEGs and cancer progression. Combined, these analyses revealed that Frondoside A involves in the regulation of multiple pathways, such as drug metabolism, cell cycle in liver cancer by inhibiting the expression of CDK1, TOP2A, CDC20, and KIF20A, and regulates protein digestion and absorption, receptor interaction in pancreatic cancer by down-regulation of ASPM, TOP2A, DLGAP5, TPX2, KIF23, MELK, LAMA3, and ANLN. While in bladder cancer, Frondoside A regulates muscle contraction, complement and coagulation cascade by increase FLNC expression. In conclusion, the present study offers valuable insights into the molecular mechanism underlying the anticancer effects of Frondoside A, and suggests that Frondoside A can be used as a functional food supplement or further developed as a natural anti-cancer drug.
Collapse
Affiliation(s)
- Guangchun Liu
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shenglin Zhang
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ruoyan Lin
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xudong Cao
- Deparment of Chemical and Biological Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Lihong Yuan
- School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
9
|
Li L, Xue Q, Zhang M, Yang Z, Wang D, Yan G, Qiao Y, Tang C, Zhang R. Upregulation of the key biomarker kinesin family member 20A (KIF20A) is associated with pulmonary artery hypertension. Genomics 2023; 115:110705. [PMID: 37703933 DOI: 10.1016/j.ygeno.2023.110705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/21/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
OBJECTIVE Pulmonary artery hypertension (PAH) is a complex, fatal disease with limited treatments. This study aimed to investigate possible key targets in PAH through bioinformatics. METHODS GSE144274 were obtained from Gene Expression Omnibus (GEO) database. Then, differentially expressed genes (DEGs) between idiopathic pulmonary hypertension (IPAH) and healthy subjects were identified and analyzed. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) were analyzed, and a protein-protein interaction (PPI) network was constructed using STRING. The hub genes were identified by MCODE method. The expression levels of hub genes were validated in vitro and in vivo models. Finally, the ROC analysis was performed based on the level of hub genes in clinical plasma samples. RESULTS A total of 363 DEGs were identified. GO analysis on these DEGs were mainly enriched in cell division, inflammatory response, among others. In the KEGG pathways analysis, DEGs mainly involved in cytokine-cytokine receptor interaction, rheumatoid arthritis, and IL-17 signaling pathways were enriched. The DEGs were analyzed with the STRING for PPI network analysis, and 62 hub genes were identified by MCODE. Finally, 6 central genes, KIF18B, SPC25, DLGAP5, KIF20A, CEP55 and ANLN, were screened out due to their novelty role in PAH. The expression of KIF20A was validated to be significantly upregulated both in the lung tissue of hypoxia-induced pulmonary hypertension (HPH) mice and proliferative PASMCs. Additionally, KIF20A levels is evelated in PAH plasma and the area under the curve (AUC) to identify PAH was 0.8591 for KIF20A. CONCLUSION The level of KIF20A elevates during the progression of PAH, which suggestes it could be a potential diagnostic and therapeutic target for the PAH.
Collapse
Affiliation(s)
- Linqing Li
- Zhongda Hospital, Southeast University, Department of Cardiology, China; Linyi People's Hospital, 210009 Nanjing, China
| | - Qi Xue
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Minhao Zhang
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Zhanneng Yang
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Dong Wang
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Gaoliang Yan
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Yong Qiao
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Chengchun Tang
- Zhongda Hospital, Southeast University, Department of Cardiology, China.
| | - Rui Zhang
- Zhongda Hospital, Southeast University, Department of Cardiology, China.
| |
Collapse
|
10
|
Ranaivoson FM, Crozet V, Benoit MPMH, Abdalla Mohammed Khalid A, Kikuti C, Sirkia H, El Marjou A, Miserey-Lenkei S, Asenjo AB, Sosa H, Schmidt CF, Rosenfeld SS, Houdusse A. Nucleotide-free structures of KIF20A illuminate atypical mechanochemistry in this kinesin-6. Open Biol 2023; 13:230122. [PMID: 37726093 PMCID: PMC10508983 DOI: 10.1098/rsob.230122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/18/2023] [Indexed: 09/21/2023] Open
Abstract
KIF20A is a critical kinesin for cell division and a promising anti-cancer drug target. The mechanisms underlying its cellular roles remain elusive. Interestingly, unusual coupling between the nucleotide- and microtubule-binding sites of this kinesin-6 has been reported, but little is known about how its divergent sequence leads to atypical motility properties. We present here the first high-resolution structure of its motor domain that delineates the highly unusual structural features of this motor, including a long L6 insertion that integrates into the core of the motor domain and that drastically affects allostery and ATPase activity. Together with the high-resolution cryo-electron microscopy microtubule-bound KIF20A structure that reveals the microtubule-binding interface, we dissect the peculiarities of the KIF20A sequence that influence its mechanochemistry, leading to low motility compared to other kinesins. Structural and functional insights from the KIF20A pre-power stroke conformation highlight the role of extended insertions in shaping the motor's mechanochemical cycle. Essential for force production and processivity is the length of the neck linker in kinesins. We highlight here the role of the sequence preceding the neck linker in controlling its backward docking and show that a neck linker four times longer than that in kinesin-1 is required for the activity of this motor.
Collapse
Affiliation(s)
- Fanomezana Moutse Ranaivoson
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| | - Vincent Crozet
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| | | | | | - Carlos Kikuti
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| | - Helena Sirkia
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| | - Ahmed El Marjou
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| | - Stéphanie Miserey-Lenkei
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| | - Ana B. Asenjo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hernando Sosa
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Christoph F. Schmidt
- Third Institute of Physics-Biophysics, Georg August University Göttingen, 37077 Göttingen, Germany
- Department of Physics and Soft Matter Center, Duke University, Durham, NC 27708, USA
| | | | - Anne Houdusse
- Structural Motility, CNRS UMR144, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, 75248 Paris, France
| |
Collapse
|
11
|
Coomar S, Mota P, Penson A, Schwaller J, Abdel-Wahab O, Gillingham D. Overlaid Transcriptional and Proteome Analyses Identify Mitotic Kinesins as Important Targets of Arylsulfonamide-Mediated RBM39 Degradation. Mol Cancer Res 2023; 21:768-778. [PMID: 37255411 PMCID: PMC10395616 DOI: 10.1158/1541-7786.mcr-22-0541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 03/16/2023] [Accepted: 05/09/2023] [Indexed: 05/14/2023]
Abstract
Certain arylsulfonamides (ArSulf) induce an interaction between the E3 ligase substrate adaptor DCAF15 and the critical splicing factor RBM39, ultimately causing its degradation. However, degradation of a splicing factor introduces complex pleiotropic effects that are difficult to untangle, since, aside from direct protein degradation, downstream transcriptional effects also influence the proteome. By overlaying transcriptional data and proteome datasets, we distinguish transcriptional from direct degradation effects, pinpointing those proteins most impacted by splicing changes. Using our workflow, we identify and validate the upregulation of the arginine-and-serine rich protein (RSRP1) and the downregulation of the key kinesin motor proteins KIF20A and KIF20B due to altered splicing in the absence of RBM39. We further show that kinesin downregulation is connected to the multinucleation phenotype observed upon RBM39 depletion by ArSulfs. Our approach should be helpful in the assessment of potential cancer drug candidates which target splicing factors. IMPLICATIONS Our approach provides a workflow for identifying and studying the most strongly modulated proteins when splicing is altered. The work also uncovers a splicing-based approach toward pharmacologic targeting of mitotic kinesins.
Collapse
Affiliation(s)
- Seemon Coomar
- Department of Chemistry, University of Basel, Basel, Switzerland
| | - Pedro Mota
- Department of Chemistry, University of Basel, Basel, Switzerland
| | - Alexander Penson
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jürg Schwaller
- Department of Biomedicine, University Children's Hospital (UKBB), University of Basel, Basel, Switzerland
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
12
|
Nishino M, Imaizumi H, Yokoyama Y, Katahira J, Kimura H, Matsuura N, Matsumura M. Histone methyltransferase SUV39H1 regulates the Golgi complex via the nuclear envelope-spanning LINC complex. PLoS One 2023; 18:e0283490. [PMID: 37437070 DOI: 10.1371/journal.pone.0283490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Cell motility is related to the higher-order structure of chromatin. Stimuli that induce cell migration change chromatin organization; such stimuli include elevated histone H3 lysine 9 trimethylation (H3K9me3). We previously showed that depletion of histone H3 lysine 9 methyltransferase, SUV39H1, suppresses directional cell migration. However, the molecular mechanism underlying this association between chromatin and cell migration remains elusive. The Golgi apparatus is a cell organelle essential for cell motility. In this study, we show that loss of H3K9 methyltransferase SUV39H1 but not SETDB1 or SETDB2 causes dispersion of the Golgi apparatus throughout the cytoplasm. The Golgi dispersion triggered by SUV39H1 depletion is independent of transcription, centrosomes, and microtubule organization, but is suppressed by depletion of any of the following three proteins: LINC complex components SUN2, nesprin-2, or microtubule plus-end-directed kinesin-like protein KIF20A. In addition, SUN2 is closely localized to H3K9me3, and SUV39H1 affects the mobility of SUN2 in the nuclear envelope. Further, inhibition of cell motility caused by SUV39H1 depletion is restored by suppression of SUN2, nesprin-2, or KIF20A. In summary, these results show the functional association between chromatin organization and cell motility via the Golgi organization regulated by the LINC complex.
Collapse
Affiliation(s)
- Miyu Nishino
- Graduate School of Health Sciences, Ehime Prefectural University of Health Sciences, Ehime, Japan
| | - Hiromasa Imaizumi
- Graduate School of Medicine and Health Science, Osaka University, Osaka, Japan
- Department of Radiological Technology, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, Okayama, Japan
| | - Yuhki Yokoyama
- Graduate School of Medicine and Health Science, Osaka University, Osaka, Japan
| | - Jun Katahira
- Laboratories of Cellular Molecular Biology, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Osaka, Japan
| | - Hiroshi Kimura
- Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Nariaki Matsuura
- Graduate School of Medicine and Health Science, Osaka University, Osaka, Japan
- Osaka International Cancer Institute, Osaka, Japan
| | - Miki Matsumura
- Graduate School of Health Sciences, Ehime Prefectural University of Health Sciences, Ehime, Japan
- Graduate School of Medicine and Health Science, Osaka University, Osaka, Japan
| |
Collapse
|
13
|
Jin Z, Peng F, Zhang C, Tao S, Xu D, Zhu Z. Expression, regulating mechanism and therapeutic target of KIF20A in multiple cancer. Heliyon 2023; 9:e13195. [PMID: 36798768 PMCID: PMC9925975 DOI: 10.1016/j.heliyon.2023.e13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Kinesin family member 20A (KIF20A) is a member of the kinesin family. It transports chromosomes during mitosis, plays a key role in cell division. Recently, studies proved that KIF20A was highly expressed in cancer. High expression of KIF20A was correlated with poor overall survival (OS). In this review, we summarized all the cancer that highly expressed KIF20A, described the role of KIF20A in cancer. We also organized phase I and phase II clinical trials of KIF20A peptides vaccine. All results indicated that KIF20A was a promising therapeutic target for multiple cancer.
Collapse
Key Words
- ATP, adenosine triphosphate
- BTC, biliary tract cancer
- CPC, chromosomal passenger complex
- CTL, cytotoxic T lymphocyte
- Cancer
- Cdk1, cyclin-dependent kinase 1
- DLG5, discs large MAGUK scaffold protein 5
- EMT, epithelial-mesenchymal transition
- Expression
- FoxM1, forkhead box protein M1
- GC, gastric cancer
- GEM, gemcitabine
- Gli2, glioma-associated oncogene 2
- HLA, human leukocyte antigen
- HNMT, head-and-neck malignant tumor
- IRF, interferon regulatory factor
- JAK, Janus kinase
- KIF20A
- KIF20A, kinesin family member 20A
- LP, long peptide
- MHC I, major histocompatibility complex I
- MKlp2, mitotic kinesin-like protein 2
- Mad2, mitotic arrest deficient 2
- OS, overall survival
- PBMC, peripheral blood mononuclear cell
- Plk1, polo-like kinase 1
- Regulating mechanisms
- Therapeutic target
- circRNA, circular RNA
- miRNA, microRNA
Collapse
Affiliation(s)
- Zheng Jin
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China
| | - Fei Peng
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Baylor College of Medicine, Houston, Texas, USA
| | - Chao Zhang
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Shuang Tao
- Department of Otorhinolaryngology Head and Neck Surgery, Longgang Central Hospital of Shenzhen, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Damo Xu
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China,State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong Province, China,Corresponding author. Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong Province, China.
| | - Zhenhua Zhu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China,Corresponding author. Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
14
|
Meng X, Li W, Yuan H, Dong W, Xiao W, Zhang X. KDELR2-KIF20A axis facilitates bladder cancer growth and metastasis by enhancing Golgi-mediated secretion. Biol Proced Online 2022; 24:12. [PMID: 36096734 PMCID: PMC9465899 DOI: 10.1186/s12575-022-00174-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Background Bladder cancer (BCa) is a fatal form of cancer worldwide associated with a poor prognosis. Identifying novel drivers of growth and metastasis hold therapeutic potential for the disease. Transport homeostasis between the endoplasmic reticulum and Golgi and the secretion of matrix metalloproteinases (MMPs) mediated by Golgi have been reported to be closely associated with tumor progression. However, to date, mechanistic studies remain limited. Results Here, we identified KDELR2 as a potential risk factor with prognostic value in patients with BCa, especially those harbouring the KDELR2 amplification. In addition, we found that KDELR2 is a regulator of BCa cell proliferation and tumorigenicity based on bioinformatic analysis with functional studies. Mechanistically, we revealed that KDELR2 could regulate the expression of KIF20A, thus stimulating the expression of MMP2, MMP9 and MKI67. Functionally, the overexpression of KDELR2 and KIF20A markedly promoted proliferation, migration, and invasion in vitro and enhanced tumor growth in vivo, while knockdown of KDELR2 and KIF20A exerted the opposite effects. And the overexpression of KDELR2 also enhanced lymph node metastasis in vivo. Conclusions Collectively, our findings clarified a hitherto unexplored mechanism of KDELR2-KIF20A axis in increasing Golgi-mediated secretion of MMPs to drive tumor progression in BCa. Supplementary Information The online version contains supplementary material available at 10.1186/s12575-022-00174-y.
Collapse
Affiliation(s)
- Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongwei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Dong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China. .,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China. .,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China. .,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
15
|
Jin Z, Tao S, Zhang C, Xu D, Zhu Z. KIF20A promotes the development of fibrosarcoma via PI3K-Akt signaling pathway. Exp Cell Res 2022; 420:113322. [PMID: 36037925 DOI: 10.1016/j.yexcr.2022.113322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 11/04/2022]
Abstract
Adult fibrosarcoma is an aggressive subtype of soft tissue sarcoma (STS), in which high expression of KIF20A indicates a poor prognosis. However, the precise role of KIF20A in fibrosarcoma progression remains unknown. In this study, we initially examined KIF20A expression and function in the human fibrosarcoma cell line HT-1080. The results showed that KIF20A was highly expressed in HT-1080, knockdown of KIF20A impaired cell proliferation, migration, invasion and induced G2/M arrest and cell apoptosis. Transcriptome study suggested that PI3K-Akt signal pathway was involved in these biological changes. We confirmed that PI3K-Akt and NF-κB signaling pathways were impaired after the down-regulation of KIF20A, which can be reversed by the Akt activator SC79 in HT-1080 in vitro. In a xenograft mouse model, knockdown of KIF20A inhibited tumor growth, Ki67 expression and liver metastasis. Taken together, our results suggested that KIF20A promoted fibrosarcoma progression via PI3K-Akt signaling pathway and might be a potential therapeutic target for fibrosarcoma.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University. Shenzhen, Guangdong Province, China
| | - Shuang Tao
- Department of Otorhinolaryngology Head and Neck Surgery, Longgang Central Hospital of Shenzhen, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Chao Zhang
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Damo Xu
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University. Shenzhen, Guangdong Province, China; State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong Province, China.
| | - Zhenhua Zhu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
16
|
Lindholm C, Batakis P, Altimiras J, Lees J. Intermittent fasting induces chronic changes in the hepatic gene expression of Red Jungle Fowl (Gallus gallus). BMC Genomics 2022; 23:304. [PMID: 35421924 PMCID: PMC9009039 DOI: 10.1186/s12864-022-08533-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/06/2022] [Indexed: 11/23/2022] Open
Abstract
Background Intermittent fasting (IF), the implementation of fasting periods of at least 12 consecutive hours on a daily to weekly basis, has received a lot of attention in recent years for imparting the life-prolonging and health-promoting effects of caloric restriction with no or only moderate actual restriction of caloric intake. IF is also widely practiced in the rearing of broiler breeders, the parent stock of meat-type chickens, who require strict feed restriction regimens to prevent the serious health problems associated with their intense appetites. Although intermittent fasting has been extensively used in this context to reduce feed competition and its resulting stress, the potential of IF in chickens as an alternative and complementary model to rodents has received less investigation. In both mammals and birds, the liver is a key component of the metabolic response to IF, responding to variations in energy balance. Here we use a microarray analysis to examine the liver transcriptomics of wild-type Red Jungle Fowl chickens fed either ad libitum, chronically restricted to around 70% of ad libitum daily or intermittently fasted (IF) on a 2:1 (2 days fed, 1 day fasted) schedule without actual caloric restriction. As red junglefowl are ancestral to domestic chicken breeds, these data serve as a baseline to which existing and future transcriptomic results from farmed birds such as broiler breeders can be compared. Results We find large effects of feeding regimen on liver transcriptomics, with most of the affected genes relating to energy metabolism. A cluster analysis shows that IF is associated with large and reciprocal changes in genes related to lipid and carbohydrate metabolism, but also chronic changes in genes related to amino acid metabolism (generally down-regulated) and cell cycle progression (generally up-regulated). The overall transcription pattern appears to be one of promoting high proliferative plasticity in response to fluctuations in available energy substrates. A small number of inflammation-related genes also show chronically changed expression profiles, as does one circadian rhythm gene. Conclusions The increase in proliferative potential suggested by the gene expression changes reported here indicates that birds and mammals respond similarly to intermittent fasting practices. Our findings therefore suggest that the health benefits of periodic caloric restriction are ubiquitous and not restricted to mammals alone. Whether a common fundamental mechanism, for example involving leptin, underpins these benefits remains to be elucidated. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08533-5.
Collapse
|
17
|
Yang Z, Wu X, Li J, Zheng Q, Niu J, Li S. CCNB2, CDC20, AURKA, TOP2A, MELK, NCAPG, KIF20A, UBE2C, PRC1, and ASPM May Be Potential Therapeutic Targets for Hepatocellular Carcinoma Using Integrated Bioinformatic Analysis. Int J Gen Med 2022; 14:10185-10194. [PMID: 34992437 PMCID: PMC8710976 DOI: 10.2147/ijgm.s341379] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/09/2021] [Indexed: 01/14/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a highly malignant, recurrent and drug-resistant tumor, and patients often lose the opportunity for surgery when they are diagnosed. Abnormal gene expression is closely related to the occurrence of HCC. The aim of the present study was to identify the differentially expressed genes (DEGs) between tumor tissue and non-tumor tissue of HCC samples in order to investigate the mechanisms of liver cancer. Methods The gene expression profile (GSE62232, GSE89377, and GSE112790) was downloaded from the Gene Expression Omnibus (GEO) and analyzed using the online tool GEO2R to identify differentially expressed genes (DEGs). Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed using the Database for Annotation, Visualization and Integrated Discovery. Protein–protein interaction (PPI) of these DEGs was analyzed based on the Search Tool for the Retrieval of Interacting Genes database and visualized by Cytoscape software. In addition, we used the online Kaplan–Meier plotter survival analysis tool to evaluate the prognostic value of hub genes expression. HPA database was used to reveal the differences in protein level of hub genes. Results A total of 50 upregulated DEGs and 122 downregulated DEGs were identified. Among them, ten hub genes with a high degree of connectivity were picked out. Overexpression of these hub genes was associated with unfavorable prognosis of HCC. Conclusion Our study suggests that CCNB2, CDC20, AURKA, TOP2A, MELK, NCAPG, KIF20A, UBE2C, PRC1, and ASPM were overexpressed in HCC compared with normal liver tissue. Overexpression of these genes was an unfavorable prognostic factor of HCC patients. Further study is needed to explore the value of them in the diagnosis and treatment of HCC. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/6-kRy19SREg
Collapse
Affiliation(s)
- Zhiqiang Yang
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xinglang Wu
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Junbo Li
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qiang Zheng
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Junwei Niu
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shengwei Li
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
18
|
Wu C, Qi X, Qiu Z, Deng G, Zhong L. Low expression of KIF20A suppresses cell proliferation, promotes chemosensitivity and is associated with better prognosis in HCC. Aging (Albany NY) 2021; 13:22148-22163. [PMID: 34491228 PMCID: PMC8507281 DOI: 10.18632/aging.203494] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022]
Abstract
This study analysed the microarray datasets from Gene Expression Omnibus (GEO) database, and aimed to identify novel potential hub genes associated with the progression of HCC via bioinformatics analysis and experimental validation. The common differentially expressed genes (DEGs) from five GEO datasets were screened using GEO2R tool. The expression and survival analysis of hub genes in HCC were performed using Gene Expression Profiling Interactive Analysis, UALCAN and Kaplan-Meier plotter tools. In vitro functional assays were used to determine the caspase-3, -9, cell proliferation and chemo-sensitivity of HCC cells. A total of 177 common DEGs were identified between normal liver and HCC tissues among these datasets. Functional enrichment and PPI network analysis identified 22 hub genes from the common DEGs. The mRNA expression of 22 hub genes was all significantly up-regulated in HCC tissues compared to that in normal liver tissues. Further survival analysis showed that 10 hub genes predicted poor prognosis of patients with HCC. More importantly, the in vitro functional studies demonstrated that KIF20A knockdown suppressed the HCC cell proliferation and promoted the chemosensitivity of HCC cells to cisplatin and sorafenib. In conclusion, the present study identified a total of 177 common DEGs among 5 GEO microarray datasets and found that 10 hub genes could predict the poor prognosis of patients with HCC using the comprehensive bioinformatics analysis. Furthermore, KIF20A silence suppressed cell proliferation and enhanced chemosensitivity in HCC cells. Further studies may be required to determine the mechanistic role of these hub genes in HCC progression.
Collapse
Affiliation(s)
- Chuanxing Wu
- Department of General Surgery, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Xiaosheng Qi
- Department of General Surgery, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Guilong Deng
- Department of General Surgery, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Lin Zhong
- Department of General Surgery, Shanghai General Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
19
|
Qiao Y, Pei Y, Luo M, Rajasekaran M, Hui KM, Chen J. Cytokinesis regulators as potential diagnostic and therapeutic biomarkers for human hepatocellular carcinoma. Exp Biol Med (Maywood) 2021; 246:1343-1354. [PMID: 33899543 DOI: 10.1177/15353702211008380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cytokinesis, the final step of mitosis, is critical for maintaining the ploidy level of cells. Cytokinesis is a complex, highly regulated process and its failure can lead to genetic instability and apoptosis, contributing to the development of cancer. Human hepatocellular carcinoma is often accompanied by a high frequency of aneuploidy and the DNA ploidy pattern observed in human hepatocellular carcinoma results mostly from impairments in cytokinesis. Many key regulators of cytokinesis are abnormally expressed in human hepatocellular carcinoma, and their expression levels are often correlated with patient prognosis. Moreover, preclinical studies have demonstrated that the inhibition of key cytokinesis regulators can suppress the growth of human hepatocellular carcinoma. Here, we provide an overview of the current understanding of the signaling networks regulating cytokinesis, the key cytokinesis regulators involved in the initiation and development of human hepatocellular carcinoma, and their applications as potential diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, P. R. China
| | - Yunxin Pei
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China
| | - Miao Luo
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China
| | - Muthukumar Rajasekaran
- Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore
| | - Kam M Hui
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore.,Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jianxiang Chen
- Pharmacy Institute and Department of Hepatology, Institute of Hepatology and Metabolic Diseases, Institute of Integrated Chinese and Western Medicine for Oncology, The affiliated Hospital of Hangzhou Normal University, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province and Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Collaborative Innovation Center of Traditional Chinese Medicines from Zhejiang Province, College of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China.,Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore
| |
Collapse
|
20
|
Saghaleyni R, Sheikh Muhammad A, Bangalore P, Nielsen J, Robinson JL. Machine learning-based investigation of the cancer protein secretory pathway. PLoS Comput Biol 2021; 17:e1008898. [PMID: 33819271 PMCID: PMC8049480 DOI: 10.1371/journal.pcbi.1008898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 04/15/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Deregulation of the protein secretory pathway (PSP) is linked to many hallmarks of cancer, such as promoting tissue invasion and modulating cell-cell signaling. The collection of secreted proteins processed by the PSP, known as the secretome, is often studied due to its potential as a reservoir of tumor biomarkers. However, there has been less focus on the protein components of the secretory machinery itself. We therefore investigated the expression changes in secretory pathway components across many different cancer types. Specifically, we implemented a dual approach involving differential expression analysis and machine learning to identify PSP genes whose expression was associated with key tumor characteristics: mutation of p53, cancer status, and tumor stage. Eight different machine learning algorithms were included in the analysis to enable comparison between methods and to focus on signals that were robust to algorithm type. The machine learning approach was validated by identifying PSP genes known to be regulated by p53, and even outperformed the differential expression analysis approach. Among the different analysis methods and cancer types, the kinesin family members KIF20A and KIF23 were consistently among the top genes associated with malignant transformation or tumor stage. However, unlike most cancer types which exhibited elevated KIF20A expression that remained relatively constant across tumor stages, renal carcinomas displayed a more gradual increase that continued with increasing disease severity. Collectively, our study demonstrates the complementary nature of a combined differential expression and machine learning approach for analyzing gene expression data, and highlights key PSP components relevant to features of tumor pathophysiology that may constitute potential therapeutic targets. The secretory pathway is a series of intracellular compartments and enzymes that process and export proteins from the cell to its surrounding environment. Dysfunction of the secretory pathway is associated with many diseases, including cancer, and therefore constitutes a potential target for novel therapeutic strategies. The large number of interacting components that comprise the secretory pathway pose a challenge when attempting to identify where the dysfunction originates or how to restore healthy function. To improve our understanding of how the secretory pathway is changed within tumors, we used gene expression data from normal tissue and tumor samples from thousands of individuals which included many different types of cancers. The data was analyzed using different machine learning algorithms which we trained to predict sample characteristics, such as disease severity. This training quantified the relative degree to which each gene was associated with the tumor characteristic, allowing us to predict which secretory pathway components were important for processes such as tumor progression—both within specific cancer types and across many different cancer types. The machine learning-based approach demonstrated excellent performance compared to traditional gene expression analysis methods and identified several secretory pathway components with strong evidence of involvement in tumor development.
Collapse
Affiliation(s)
- Rasool Saghaleyni
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Azam Sheikh Muhammad
- Department of Computer Science and Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Wallenberg Center for Protein Research, Chalmers University of Technology, Gothenburg, Sweden
- BioInnovation Institute, Copenhagen, Denmark
| | - Jonathan L. Robinson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Wallenberg Center for Protein Research, Chalmers University of Technology, Gothenburg, Sweden
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Gothenburg, Sweden
- * E-mail:
| |
Collapse
|
21
|
Fang ZX, Niu JJ, Liu PG, Lin Y. Genome-wide long noncoding RNA and mRNA expression profiles demonstrate associations between exposure to inorganic elements and the risk of developing hepatocellular carcinoma. BMC Med Genomics 2021; 14:85. [PMID: 33736645 PMCID: PMC7977256 DOI: 10.1186/s12920-021-00927-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 02/28/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are closely associated with the development of hepatocellular carcinoma (HCC). The present study conducted a genome-wide microarray analysis and qPCR validation to obtain comprehensive insights into this issue. METHODS Thirty male HCC patients with chronic HBV infection were included in the present study. Primary HCC tissue and normal tissue were collected. Double-stranded complementary DNA synthesized from 10 pairs of samples was labeled and hybridized to a microarray chip. Further analyses, such as hierarchical clustering, gene ontology (GO) and pathway analyses, were performed. In addition, qPCR validation was performed on tissue samples and additional serum samples. RESULTS The microarray analysis identified 946 upregulated and 571 downregulated lncRNAs and 1720 upregulated and 1106 downregulated mRNAs. Among these RNAs, ENST00000583827.1 (fold change: 21) and uc010isf.1 (fold change: 18) were the most over- and underexpressed lncRNAs in the HCC tissues, respectively. For the mRNAs, KIF20A (fold change: 26) and HEPACAM (fold change: 50) were the most over- and underexpressed in the HCC tissues, respectively. The GO analysis demonstrated that the most differentially expressed mRNAs were related to the response of metal ions. The pathway analysis also suggested that the most enriched pathway was mineral absorption. CONCLUSIONS The subsequent qPCR validation exhibited high consistency with the microarray analysis, except for three lncRNAs. The qPCR analysis also demonstrated that TCONS_00008984 had a 767-fold overexpression level in HCC tissues when compared with normal tissues, and this finding was confirmed in the serum samples; therefore, TCONS_00008984 has the potential to serve as a diagnostic marker or prognostic indicator. The GO and pathway analyses indicated that exposure to inorganic elements may be involved in HCC risk.
Collapse
Affiliation(s)
- Zan-Xi Fang
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, 209 Hubin South Road, Xiamen, 361004, Fujian Province, China
| | - Jian-Jun Niu
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, 209 Hubin South Road, Xiamen, 361004, Fujian Province, China
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, China
| | - Ping-Guo Liu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Xiamen, China
| | - Yong Lin
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, 209 Hubin South Road, Xiamen, 361004, Fujian Province, China.
- Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
22
|
MDM4 inhibition: a novel therapeutic strategy to reactivate p53 in hepatoblastoma. Sci Rep 2021; 11:2967. [PMID: 33536467 PMCID: PMC7859402 DOI: 10.1038/s41598-021-82542-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatoblastoma (HB) is the most common pediatric liver malignancy. High-risk patients have poor survival, and current chemotherapies are associated with significant toxicities. Targeted therapies are needed to improve outcomes and patient quality of life. Most HB cases are TP53 wild-type; therefore, we hypothesized that targeting the p53 regulator Murine double minute 4 (MDM4) to reactivate p53 signaling may show efficacy. MDM4 expression was elevated in HB patient samples, and increased expression was strongly correlated with decreased expression of p53 target genes. Treatment with NSC207895 (XI-006), which inhibits MDM4 expression, or ATSP-7041, a stapled peptide dual inhibitor of MDM2 and MDM4, showed significant cytotoxic and antiproliferative effects in HB cells. Similar phenotypes were seen with short hairpin RNA (shRNA)-mediated inhibition of MDM4. Both NSC207895 and ATSP-7041 caused significant upregulation of p53 targets in HB cells. Knocking-down TP53 with shRNA or overexpressing MDM4 led to resistance to NSC207895-mediated cytotoxicity, suggesting that this phenotype is dependent on the MDM4-p53 axis. MDM4 inhibition also showed efficacy in a murine model of HB with significantly decreased tumor weight and increased apoptosis observed in the treatment group. This study demonstrates that inhibition of MDM4 is efficacious in HB by upregulating p53 tumor suppressor signaling.
Collapse
|
23
|
Zeng XC, Zhang L, Liao WJ, Ao L, Lin ZM, Kang W, Chen WN, Lin X. Screening and Identification of Potential Biomarkers in Hepatitis B Virus-Related Hepatocellular Carcinoma by Bioinformatics Analysis. Front Genet 2020; 11:555537. [PMID: 33193629 PMCID: PMC7556301 DOI: 10.3389/fgene.2020.555537] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers globally. Hepatitis B virus (HBV) infection might cause chronic hepatitis and cirrhosis, leading to HCC. To screen prognostic genes and therapeutic targets for HCC by bioinformatics analysis and determine the mechanisms underlying HBV-related HCC, three high-throughput RNA-seq based raw datasets, namely GSE25599, GSE77509, and GSE94660, were obtained from the Gene Expression Omnibus database, and one RNA-seq raw dataset was acquired from The Cancer Genome Atlas (TCGA). Overall, 103 genes were up-regulated and 127 were down-regulated. A protein–protein interaction (PPI) network was established using Cytoscape software, and 12 pivotal genes were selected as hub genes. The 230 differentially expressed genes and 12 hub genes were subjected to functional and pathway enrichment analyses, and the results suggested that cell cycle, nuclear division, mitotic nuclear division, oocyte meiosis, retinol metabolism, and p53 signaling-related pathways play important roles in HBV-related HCC progression. Further, among the 12 hub genes, kinesin family member 11 (KIF11), TPX2 microtubule nucleation factor (TPX2), kinesin family member 20A (KIF20A), and cyclin B2 (CCNB2) were identified as independent prognostic genes by survival analysis and univariate and multivariate Cox regression analysis. These four genes showed higher expression levels in HCC than in normal tissue samples, as identified upon analyses with Oncomine. In addition, in comparison with normal tissues, the expression levels of KIF11, TPX2, KIF20A, and CCNB2 were higher in HBV-related HCC than in HCV-related HCC tissues. In conclusion, our results suggest that KIF11, TPX2, KIF20A, and CCNB2 might be involved in the carcinogenesis and development of HBV-related HCC. They can thus be used as independent prognostic genes and novel biomarkers for the diagnosis of HBV-related HCC and development of pertinent therapeutic strategies.
Collapse
Affiliation(s)
- Xian-Chang Zeng
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Lu Zhang
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wen-Jun Liao
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Lu Ao
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ze-Man Lin
- Fujian Key Laboratory of Medical Bioinformatics, Department of Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wen Kang
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wan-Nan Chen
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
24
|
Li Y, Wei Z, Huang S, Yang B. mRNA expression and DNA methylation analysis of the inhibitory mechanism of H 2O 2 on the proliferation of A549 cells. Oncol Lett 2020; 20:288. [PMID: 33014166 PMCID: PMC7520746 DOI: 10.3892/ol.2020.12151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/18/2020] [Indexed: 01/29/2023] Open
Abstract
Reactive oxygen species, particularly hydrogen peroxide (H2O2), can induce proliferation inhibition and death of A549 cells via oxidative stress. Oxidative stress has effect on DNA methylation. Oxidative stress and DNA methylation feature a common denominator: The one carbon cycle. To explore the inhibitory mechanism of H2O2 on the proliferation of lung cancer cells, the present study analysed the mRNA expression and methylation profiles in A549 cells treated with H2O2 for 24 h, as adenocarcinoma is the most common pathological type of lung cancer. The DNA methylation profile was constructed using reduced representation bisulphite sequencing, which identified 29,755 differentially methylated sites (15,365 upregulated and 14,390 downregulated), and 1,575 differentially methylated regions located in the gene promoters were identified using the methylKit. Analysis of the assocaition between gene expression and methylation levels revealed that several genes were downregulated and hypermethylated, including cyclin-dependent kinase inhibitor 3, denticleless E3 ubiquitin protein ligase homolog, centromere protein (CENP)F, kinesin family member (KIF)20A, CENPA, KIF11, PCNA clamp-associated factor and GINS complex subunit 2, which may be involved in the inhibitory process of H2O2 on the proliferation of A549 cells.
Collapse
Affiliation(s)
- Yepeng Li
- Department of Oncology, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Zhongheng Wei
- Department of Oncology, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Shiqing Huang
- Department of Oncology, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| | - Bo Yang
- Key Laboratory of Guangxi College and Universities, Biomedical Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region 533000, P.R. China
| |
Collapse
|
25
|
KIF20A Predicts Poor Survival of Patients and Promotes Colorectal Cancer Tumor Progression through the JAK/STAT3 Signaling Pathway. DISEASE MARKERS 2020; 2020:2032679. [PMID: 32695240 PMCID: PMC7368235 DOI: 10.1155/2020/2032679] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/14/2020] [Accepted: 06/13/2020] [Indexed: 12/24/2022]
Abstract
Kinesin family member 20A (KIF20A) has been recently reported to be upregulated and associated with increased invasiveness and metastasis in several malignancies. However, the role of KIF20A in colorectal cancer (CRC) is still unclear. This study is aimed at investigating the potential roles of KIF20A in the development of CRC. The results of bioinformatics analysis, immunohistochemical staining, and Western blot analysis showed that KIF20A was overexpressed in CRC tissues compared with adjacent normal tissues. High expression of KIF20A in CRC tissues was associated with depth of invasion, lymphatic node metastasis, distant metastasis, and TNM stage. Moreover, the Kaplan-Meier survival analysis showed that CRC patients with high KIF20A expression had poor prognoses. Cox regression analysis revealed that KIF20A was an independent prognostic factor in patients with CRC. Further studies suggested that knockdown of KIF20A was able to reduce cell proliferation and migration by inhibiting the JAK/STAT3 pathway. Taken together, we propose that KIF20A plays a critical role in the tumorigenesis and tumor progression of colorectal cancer and could represent a potential therapeutic target for CRC.
Collapse
|
26
|
Ouyang G, Yi B, Pan G, Chen X. A robust twelve-gene signature for prognosis prediction of hepatocellular carcinoma. Cancer Cell Int 2020; 20:207. [PMID: 32514252 PMCID: PMC7268417 DOI: 10.1186/s12935-020-01294-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Background The prognosis of hepatocellular carcinoma (HCC) patients remains poor. Identifying prognostic markers to stratify HCC patients might help to improve their outcomes. Methods Six gene expression profiles (GSE121248, GSE84402, GSE65372, GSE51401, GSE45267 and GSE14520) were obtained for differentially expressed genes (DEGs) analysis between HCC tissues and non-tumor tissues. To identify the prognostic genes and establish risk score model, univariable Cox regression survival analysis and Lasso-penalized Cox regression analysis were performed based on the integrated DEGs by robust rank aggregation method. Then Kaplan-Meier and time-dependent receiver operating characteristic (ROC) curves were generated to validate the prognostic performance of risk score in training datasets and validation datasets. Multivariable Cox regression analysis was used to identify independent prognostic factors in liver cancer. A prognostic nomogram was constructed based on The Cancer Genome Atlas (TCGA) dataset. Finally, the correlation between DNA methylation and prognosis-related genes was analyzed. Results A twelve-gene signature including SPP1, KIF20A, HMMR, TPX2, LAPTM4B, TTK, MAGEA6, ANX10, LECT2, CYP2C9, RDH16 and LCAT was identified, and risk score was calculated by corresponding coefficients. The risk score model showed a strong diagnosis performance to distinguish HCC from normal samples. The HCC patients were stratified into high-risk and low-risk group based on the cutoff value of risk score. The Kaplan-Meier survival curves revealed significantly favorable overall survival in groups with lower risk score (P < 0.0001). Time-dependent ROC analysis showed well prognostic performance of the twelve-gene signature, which was comparable or superior to AJCC stage at predicting 1-, 3-, and 5-year overall survival. In addition, the twelve-gene signature was independent with other clinical factors and performed better in predicting overall survival after combining with age and AJCC stage by nomogram. Moreover, most of the prognostic twelve genes were negatively correlated with DNA methylation in HCC tissues, which SPP1 and LCAT were identified as the DNA methylation-driven genes. Conclusions We identified a twelve-gene signature as a robust marker with great potential for clinical application in risk stratification and overall survival prediction in HCC patients.
Collapse
Affiliation(s)
- Guoqing Ouyang
- Department of Hepatobiliary Surgery, Liuzhou People's Hospital, Liuzhou, China
| | - Bin Yi
- Department of Cardio-Vascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangdong Pan
- Department of Hepatobiliary Surgery, Liuzhou People's Hospital, Liuzhou, China
| | - Xiang Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Xie F, He C, Gao S, Yang Z, Li L, Qiao L, Fang L. KIF20A silence inhibits the migration, invasion and proliferation of non-small cell lung cancer and regulates the JNK pathway. Clin Exp Pharmacol Physiol 2020; 47:135-142. [PMID: 31557334 DOI: 10.1111/1440-1681.13183] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022]
Abstract
An increasing number of studies have shown that kinesin family member 20A (KIF20A) was overexpressed in several types of cancer, and its overexpression correlated with the oncogenesis and prognosis of cancers. However, little is known about the roles of KIF20A in human non-small cell lung cancer (NSCLC). Thus, the aim of the present study was to demonstrate the expression of KIF20A in human NSCLC and reveal its biological functions and the underlying mechanisms. qRT-PCR, western blot and immunohistochemistry were used to assess the expression of NSCLC patient specimens and NSCLC cell lines. The functions of KIF20A in migration and invasion were determined using Transwell assay. Cell proliferation capacity was performed by CKK-8 assay. We demonstrated that KIF20A was overexpressed in NSCLC specimens compared with the adjacent non-tumorous specimens, and high expression of KIF20A was associated with clinical stage and metastasis in NSCLC. Decreased expression of KIF20A inhibited NSCLC cells migration, invasion and proliferation. Most importantly, further experiments demonstrated that decreased the expression of KLF20A significantly downregulated expression of p-JNK and MMP7, which indicated that knockdown of KIF20A alters lung cancer cell phenotype and regulates JNK pathways. These results suggest that KIF20A may act as a putative oncogene and a potential therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Feng Xie
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| | - Chengyan He
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| | - Shen Gao
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| | - Zhaowei Yang
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| | - Lihong Li
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| | - Lu Qiao
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| | - Ling Fang
- China-Japan Union Hospital of Jilin Universtity, Chang chun, China
| |
Collapse
|
28
|
Kinesin-6 family motor KIF20A regulates central spindle assembly and acrosome biogenesis in mouse spermatogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118636. [PMID: 31884069 DOI: 10.1016/j.bbamcr.2019.118636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/15/2019] [Accepted: 12/23/2019] [Indexed: 11/21/2022]
Abstract
Kinesin-6 KIF20A is essential for microtubule organization and central spindle assembly during cytokinesis. However, the functions of KIF20A in meiotic division and spermatogenesis remain elusive. Here, we report that kinesin-6 KIF20A locates at the microtubules in mouse spermatogenic cells and co-localizes with the spindle midzone and midbody. We demonstrate that central spindle organization and chromosomal stability are regulated by KIF20A in male meiotic division. KIF20A inhibition leads to the defects in central spindle assembly and cytokinetic abscission, and finally results in the increase of aneuploid cells and the alteration of cell populations in the spermatogenic cells. Furthermore, we have revealed that kinesin-6 KIF20A is associated with the formation and maturation of the acrosomes during spermatogenesis. Our findings have identified the specific roles of KIF20A in central spindle organization in meiotic division.
Collapse
|
29
|
Mandal K, Pogoda K, Nandi S, Mathieu S, Kasri A, Klein E, Radvanyi F, Goud B, Janmey PA, Manneville JB. Role of a Kinesin Motor in Cancer Cell Mechanics. NANO LETTERS 2019; 19:7691-7702. [PMID: 31565944 PMCID: PMC7737127 DOI: 10.1021/acs.nanolett.9b02592] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Molecular motors play important roles in force generation, migration, and intracellular trafficking. Changes in specific motor activities are altered in numerous diseases. KIF20A, a motor protein of the kinesin-6 family, is overexpressed in bladder cancer, and KIF20A levels correlate negatively with clinical outcomes. We report here a new role for the KIF20A kinesin motor protein in intracellular mechanics. Using optical tweezers to probe intracellular mechanics and surface AFM to probe cortical mechanics, we first confirm that bladder urothelial cells soften with an increasing cancer grade. We then show that inhibiting KIF20A makes the intracellular environment softer for both high- and low-grade bladder cancer cells. Upon inhibition of KIF20A, cortical stiffness also decreases in lower grade cells, while it surprisingly increases in higher grade malignant cells. Changes in cortical stiffness correlate with the interaction of KIF20A with myosin IIA. Moreover, KIF20A inhibition negatively regulates bladder cancer cell motility irrespective of the underlying substrate stiffness. Our results reveal a central role for a microtubule motor in cell mechanics and migration in the context of bladder cancer.
Collapse
Affiliation(s)
- Kalpana Mandal
- Institute for Medicine and Engineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Katarzyna Pogoda
- Institute for Medicine and Engineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
- Institute of Nuclear Physics , Polish Academy of Sciences , PL-31342 Krakow 31-342 , Poland
| | - Satabdi Nandi
- School of Veterinary Medicine , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
- Laboratory of Molecular Biology and Immunology , National Institute on Aging , Baltimore , Maryland 21224 , United States
| | - Samuel Mathieu
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| | - Amal Kasri
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
- ICM Brain and Spine Institute , Pitié Salpêtrière Hospital , 47-83 Boulevard de l'Hôpital , Paris 75013 , France
| | - Eric Klein
- Department of Biology , Rutgers University-Camden Waterfront Tech Center , Camden , New Jersey 08103 , United States
| | - François Radvanyi
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| | - Bruno Goud
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| | - Paul A Janmey
- Institute for Medicine and Engineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
- Departments of Physiology and Physics & Astronomy , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Jean-Baptiste Manneville
- Institut Curie, PSL Research University, CNRS, UMR 144 , 26 rue d'Ulm , Paris Cedex 05 75248 , France
| |
Collapse
|
30
|
Li X, Shu K, Wang Z, Ding D. Prognostic significance of KIF2A and KIF20A expression in human cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2019; 98:e18040. [PMID: 31725680 PMCID: PMC6867763 DOI: 10.1097/md.0000000000018040] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The kinesin family (KIF) is reported to be aberrantly expressed and significantly correlated with survival outcomes in patients with various cancers. This meta-analysis was carried out to quantitatively evaluate the prognostic values of partial KIF members in cancer patients. METHODS Two well-known KIF members, KIF2A and KIF20A, were investigated to evaluate their potential values as novel prognostic biomarkers in human cancer. A comprehensive literature search was carried out of the PubMed, EMBASE, Cochrane Library, and Web of Science databases up to April 2019. Pooled hazard ratios (HRs) and odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to assess the association of KIF2A and KIF20A expression with overall survival (OS) and clinicopathological parameters. RESULTS Twenty-five studies involving 7262 patients were finally incorporated, including nine about KIF2A and sixteen about KIF20A. Our results indicated that patients with high expression of KIF2 and KIF20A tended to have shorter OS than those with low expression (HR = 2.23, 95% CI = 1.87-2.65, P < .001; HR = 1.77, 95% CI = 1.57-1.99, P < .001, respectively). Moreover, high expression of these 2 KIF members was significantly associated with advanced clinical stage (OR = 1.98, 95% CI: 1.57-2.50, P < .001; OR = 2.63, 95% CI: 2.03-3.41, P < .001, respectively), positive lymph node metastasis (OR = 2.32, 95% CI: 1.65-3.27, P < .001; OR = 2.13, 95% CI: 1.59-2.83, P < .001, respectively), and distant metastasis (OR = 2.20, 95% CI: 1.21-3.99, P = .010; OR = 5.25, 95% CI: 2.82-9.77, P < .001, respectively); only high KIF20A expression was related to poor differentiation grade (OR = 1.82, 95% CI: 1.09-3.07, P = .023). CONCLUSIONS High expression of KIF2 and KIF20A in human cancer was significantly correlated with worse prognosis and unfavorable clinicopathological features, suggesting that these 2 KIF members can be used as prognostic biomarkers for different types of tumors. PROSPERO REGISTRATION NUMBER CRD42019134928.
Collapse
Affiliation(s)
- Xing Li
- Department of Urology, People's Hospital of Zhengzhou University
| | - Kunpeng Shu
- Department of Urology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Zhifeng Wang
- Department of Urology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Degang Ding
- Department of Urology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| |
Collapse
|
31
|
Hasan MS, Wu X, Zhang L. Uncovering missed indels by leveraging unmapped reads. Sci Rep 2019; 9:11093. [PMID: 31366961 PMCID: PMC6668410 DOI: 10.1038/s41598-019-47405-z] [Citation(s) in RCA: 6] [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: 03/22/2019] [Accepted: 07/12/2019] [Indexed: 02/08/2023] Open
Abstract
In current practice, Next Generation Sequencing (NGS) applications start with mapping/aligning short reads to the reference genome, with the aim of identifying genetic variants. Although existing alignment tools have shown great accuracy in mapping short reads to the reference genome, a significant number of short reads still remain unmapped and are often excluded from downstream analyses thereby causing nonnegligible information loss in the subsequent variant calling procedure. This paper describes Genesis-indel, a computational pipeline that explores the unmapped reads to identify novel indels that are initially missed in the original procedure. Genesis-indel is applied to the unmapped reads of 30 breast cancer patients from TCGA. Results show that the unmapped reads are conserved between the two subtypes of breast cancer investigated in this study and might contribute to the divergence between the subtypes. Genesis-indel identifies 72,997 novel high-quality indels previously not found, among which 16,141 have not been annotated in the widely used mutation database. Statistical analysis of these indels shows significant enrichment of indels residing in oncogenes and tumour suppressor genes. Functional annotation further reveals that these indels are strongly correlated with pathways of cancer and can have high to moderate impact on protein functions. Additionally, some of the indels overlap with the genes that do not have any indel mutations called from the originally mapped reads but have been shown to contribute to the tumorigenesis in multiple carcinomas, further emphasizing the importance of rescuing indels hidden in the unmapped reads in cancer and disease studies.
Collapse
Affiliation(s)
| | - Xiaowei Wu
- Department of Statistics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Liqing Zhang
- Department of Computer Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
32
|
Wu WD, Yu KW, Zhong N, Xiao Y, She ZY. Roles and mechanisms of Kinesin-6 KIF20A in spindle organization during cell division. Eur J Cell Biol 2019; 98:74-80. [DOI: 10.1016/j.ejcb.2018.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
|
33
|
KIF20A Affects the Prognosis of Bladder Cancer by Promoting the Proliferation and Metastasis of Bladder Cancer Cells. DISEASE MARKERS 2019; 2019:4863182. [PMID: 31093305 PMCID: PMC6481133 DOI: 10.1155/2019/4863182] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/23/2019] [Accepted: 03/05/2019] [Indexed: 01/16/2023]
Abstract
Objective To investigate the expression of kinesin family member 20A (KIF20A) in bladder cancer, the effect of KIF20A on the proliferation and metastasis of bladder cancer cells, and the effect of KIF20A expression on the prognosis of bladder cancer patients. Methods Bladder cancer tissue and its adjacent tissues were collected from tumour patients. The mRNA and protein expression levels of KIF20A in the tissue samples were detected by qRT-PCR and western blot. Immunohistochemical (IHC) staining was used to identify the expression and distribution of KIF20A proteins in the tissue samples. The relationship between the KIF20A expression and the clinical pathology of bladder cancer was analysed. The effect of the differential expression of KIF20A on the prognosis of patients with bladder cancer was analysed by the TCGA database. The plasmid was transfected into the bladder cell lines T24 and 5637 to construct two stable cell lines with knocked down KIF20A. The effect of KIF20A expression on the proliferation and invasion of T24 and 5637 bladder cells was explored in vitro using the abovementioned stable cell lines. The effect of the KIF20A expression on the proliferation of bladder cancer cells was evaluated by a mouse xenograft model. Results The expression of KIF20A was significantly higher in the bladder cancer tissues than in the adjacent control tissues. The expression of KIF20A was significantly associated with the degree of pathological differentiation of bladder cancer. Patients with a higher expression of KIF20A had a higher tumour grade and a more advanced stage. The mean survival of patients with a high KIF20A expression was significantly lower than the mean survival of patients with a low KIF20A expression. The in vitro experiments demonstrated that the knockdown of KIF20A significantly inhibited T24 and 5637 cell proliferation and invasion. The in vivo experiments showed that the knockdown of KIF20A significantly inhibited the proliferation of the bladder tumours. Conclusion KIF20A promotes the proliferation and metastasis of bladder cancer cells. Bladder cancer patients with a high KIF20A expression have a worse tumour differentiation and a poor prognosis. KIF20A may become an independent factor that affects the prognosis of bladder cancer patients and a therapeutic target for bladder cancer.
Collapse
|
34
|
Lactate Activates the E2F Pathway to Promote Cell Motility by Up-Regulating Microtubule Modulating Genes. Cancers (Basel) 2019; 11:cancers11030274. [PMID: 30813560 PMCID: PMC6468617 DOI: 10.3390/cancers11030274] [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: 01/28/2019] [Revised: 02/21/2019] [Accepted: 02/21/2019] [Indexed: 12/29/2022] Open
Abstract
Excess lactate production due to enhanced aerobic glycolysis is characteristic of malignant cancers, which is also intimately associated with poor cancer prognoses. Although tumor-associated lactate contributes to all major steps in carcinogenesis, its action mechanism remains obscure. To understand the molecular mechanism of the lactate-induced tumor metastatic process, we identified an array of lactate-responsive genes via transcriptome analysis of a metformin-induced hyper-glycolytic liver cancer model. Gene set enrichment analysis suggested E2F-RB pathway as the dominant regulator of the lactate-induced gene expression. We experimentally verified that lactate indeed activates E2F-mediated transcription by promoting E2F1 protein accumulation through a posttranscriptional mechanism. Literature-based analysis of target pathways potentially modulated by 136 top-ranked genes indicated that genes functioning in cell-cell or cell-matrix communications dominate the lactate-induced gene expression. Especially, those regulating microtubule functions, including a group of kinesin family members, were significantly up-regulated in lactate- and E2F1-dependent manners. Depletion of E2F1 or kinesins (KIF2C, KIF18B, KIF20A) led to deformation of microtubule structures, impairing cell motility as much as the deficit in lactate production. These results indicate that E2F pathway activation by tumor-associated lactate and subsequent transcriptional activation of microtubule functions play crucial roles in tumor metastasis, providing mechanistic clues to cell motility-directed anti-cancer strategies.
Collapse
|
35
|
Song Z, Huang Y, Zhao Y, Ruan H, Yang H, Cao Q, Liu D, Zhang X, Chen K. The Identification of Potential Biomarkers and Biological Pathways in Prostate Cancer. J Cancer 2019; 10:1398-1408. [PMID: 31031850 PMCID: PMC6485223 DOI: 10.7150/jca.29571] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022] Open
Abstract
Purpose: The present study aims to explore the potential mechanisms contributing to prostate cancer (PCa), screen the hub genes, and identify potential biomarkers and correlated pathways of PCa progression. Methods: The PCa gene expression profile GSE3325 was operated to analyze the differentially expressed genes (DEGs). DAVID was used to evaluate Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. A protein-protein interaction (PPI) network was constructed to visualize interactions of the hub genes. The prognostic and diagnostic analysis of these hub genes was carried out to evaluate their potential effects on PCa. Results: A total of 847 DEGs were identified (427 upregulated genes and 420 downregulated genes). Meanwhile, top15 hub genes were showed. GO analysis displayed that the DEGs were mainly enriched in cell cycle, DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrest and proteinaceous extracellular matrix. KEGG analysis indicated the DEGs were enriched in the p53 signaling pathway and cell cycle pathway. The GO and KEGG enrichment analyses for the DEGs disclosed important biological features of PCa. PPI network showed the interaction of top 15 hub genes. Gene Set Enrichment Analysis (GSEA) revealed that some of the hub genes were associated with biochemical recurrence (BCR) and metastasis of PCa. Some top hub genes were distinctive and new discoveries compared with that of the existing associated researches. Conclusions: Our analysis revealed that the changes of cell cycle and p53 signaling pathway are two major signatures of PCa. CENPA, KIF20A and CDCA8 might promote the tumorigenesis and progression of PCa, especially in BCR and metastasis, which could be novel therapeutic targets and biomarkers for diagnosis, prognosis of PCa.
Collapse
Affiliation(s)
- Zhengshuai Song
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Urology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology
| | - Yu Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ye Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qi Cao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
36
|
Abstract
Whole-genome and centrosome duplication as a consequence of cytokinesis failure can drive tumorigenesis in experimental model systems. However, whether cytokinesis failure is in fact an important cause of human cancers has remained unclear. In this Review, we summarize evidence that whole-genome-doubling events are frequently observed in human cancers and discuss the contribution that cytokinesis defects can make to tumorigenesis. We provide an overview of the potential causes of cytokinesis failure and discuss how tetraploid cells that are generated through cytokinesis defects are used in cancer as a transitory state on the route to aneuploidy. Finally, we discuss how cytokinesis defects can facilitate genetic diversification within the tumour to promote cancer development and could constitute the path of least resistance in tumour evolution.
Collapse
Affiliation(s)
- Susanne M A Lens
- Oncode Institute, Utrecht, Netherlands.
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.
| | - René H Medema
- Oncode Institute, Utrecht, Netherlands.
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, Amsterdam, Netherlands.
| |
Collapse
|
37
|
Yuan B, Chen Y, Wu Z, Zhang L, Zhuang Y, Zhao X, Niu H, Cheng JCH, Zeng Z. Proteomic Profiling of Human Hepatic Stellate Cell Line LX2 Responses to Irradiation and TGF-β1. J Proteome Res 2018; 18:508-521. [PMID: 30489086 DOI: 10.1021/acs.jproteome.8b00814] [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] [Indexed: 12/15/2022]
Abstract
Hepatic stellate cells (HSCs) are the main target of radiation damage and primarily contribute to the development of radiation-induced liver fibrosis. However, the molecular events underlying the radiation-induced activation of HSCs are not fully elucidated. In the present study, human HSC line LX2 was treated with X-ray irradiation and/or TGF-β1, and profibrogenic molecules were evaluated. The iTRAQ LC-MS/MS technology was performed to identify global protein expression profiles in LX2 following exposure to different stimuli. Irradiation or TGF-β1 alone increased expression of α-SMA, collagen 1, CTGF, PAI-1, and fibronectin. Irradiation and TGF-β1 cooperatively induced expression of these profibrotic markers. In total, 102, 137, 155 dysregulated proteins were identified in LX2 cell samples affected by irradiation, TGF-β1, or cotreatment, respectively. Bioinformatic analyses showed that the three differentially expressed protein sets were commonly associated with cell cycle and protein processing in endoplasmic reticulum. The expression of a set of proteins was properly validated: CDC20, PRC1, KIF20A, CCNB1, SHCBP, TACC3 were upregulated upon irradiation or irradiation and TGF-β1 costimulation, whereas SPARC and THBS1 were elevated by TGF-β1 or TGF-β1 plus irradiation treatment. Furthermore, CDC20 inhibition suppressed expression of profibrotic markers in irradiated and TGF-β1-stimulated LX2 cells. Detailed data on potential molecular mechanisms causing the radiation-induced HSC activation presented here would be instrumental in developing radiotherapy strategies that minimize radiation-induced liver fibrosis.
Collapse
Affiliation(s)
- Baoying Yuan
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Yuhan Chen
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China.,Department of Radiation Oncology, Nanfang Hospital , Southern Medical University , Guangzhou 510515 , China
| | - Zhifeng Wu
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Li Zhang
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Yuan Zhuang
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Xiaomei Zhao
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Hao Niu
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| | - Jason Chia-Hsien Cheng
- Division of Radiation Oncology, Departments of Oncology , National Taiwan University Hospital , Taipei 100 , Taiwan
| | - Zhaochong Zeng
- Department of Radiation Oncology, Zhongshan Hospital , Fudan University , Shanghai 200032 , China
| |
Collapse
|
38
|
Sheng Y, Wang W, Hong B, Jiang X, Sun R, Yan Q, Zhang S, Lu M, Wang S, Zhang Z, Lin W, Li Y. Upregulation of KIF20A correlates with poor prognosis in gastric cancer. Cancer Manag Res 2018; 10:6205-6216. [PMID: 30538567 PMCID: PMC6260125 DOI: 10.2147/cmar.s176147] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background KIF20A is well known as one of the key proteins in mitosis. Recently, a number of studies illustrated that KIF20A might function as an oncogene in some carcinomas. However, its expression levels and clinical value remained unclear in gastric cancer (GC). Patients and methods In this study, we investigated the expression of KIF20A in samples from GC patients and cell lines by quantitative real-time PCR and Western blot. The function of KIF20A in cell proliferation of GC cell lines was examined via cell viability and colony formation assays. Immunohistochemistry assay based on a tissue microarray consisting of 146 cases was performed to evaluate the prognostic value of KIF20A. The overall survival rate of 122 GC patients based on KIF20A expression was analyzed as well. Finally, using KIF20A inhibitor, genistein, and combining it with cisplatin or fluorouracil, the antitumor effects were studied. Results Most GC samples (56.76%) showed higher KIF20A expression level compared to their corresponding normal specimens, which demonstrated the potential oncogenic role of KIF20A in GC. The functional studies elucidated the essential role of KIF20A in GC cell proliferation. Besides, tissue microarray result showed that the expression level of KIF20A was significantly related to the histological grades (P=0.036). Furthermore, we found the expression of KIF20A was related to poor overall survival rate, which is coincident with the results from Kaplan–Meier plotter database. In addition, we found that a KIF20A inhibitor, genistein, could enhance the antitumor activity of cisplatin and fluorouracil, which might be considered as a chemosensitive agent in GC. Conclusion KIF20A can promote cell proliferation in GC, which might be used as an independent prognostic factor and a potential therapeutic target.
Collapse
Affiliation(s)
- Yi Sheng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Wei Wang
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China,
| | - Bo Hong
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China,
| | - Xingwang Jiang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Ruochuan Sun
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Qiang Yan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Shangxin Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Mingdian Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Shengyi Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Zhen Zhang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| | - Wenchu Lin
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, China,
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China,
| |
Collapse
|
39
|
Li H, Zhang W, Sun X, Chen J, Li Y, Niu C, Xu B, Zhang Y. Overexpression of kinesin family member 20A is associated with unfavorable clinical outcome and tumor progression in epithelial ovarian cancer. Cancer Manag Res 2018; 10:3433-3450. [PMID: 30254487 PMCID: PMC6140728 DOI: 10.2147/cmar.s169214] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background KIF20A plays an indispensable role in cytokinesis regulation, which is important for tumor proliferation and growth. Recently, the oncogenic role of KIF20A has been well documented in several cancers. However, its clinical role in epithelial ovarian cancer (EOC) remains not reported yet. We investigated its expression and its role in promoting invasion and chemoresistance in EOC cells. Patients and methods KIF20A transcription and translation levels were investigated in normal ovarian epithelial cell, ovarian cancer cells, and 10 pairs of fresh EOC tissues and adjacent normal ovarian tissues by real-time quantitative polymerase chain reaction and Western blots. Moreover, KIF20A protein level was also examined by immunohistochemistry in 150 EOC tissues. The correlation between KIF20A expression and clinical variables was analyzed by statistical methods. We also used wound healing assay, transwell assay MTT, and Annexin V/PI to explore KIF20A functions. Results KIF20A expression was obviously elevated at both mRNA and protein levels in EOC cell lines and clinical cancer tissues compared with normal ovarian epithelial cell and adjacent normal ovarian tissues. KIF20A protein expression was highly correlated with International Federation of Gynecology and Obstetrics stage (P=0.008), lymph node metastasis (P=0.002), intraperitoneal metastasis (P<0.001), vital status at last follow-up (P<0.001), intraperitoneal recurrence (P=0.030), tumor recurrence (P=0.005), drug resistance (P=0.013), and ascites with tumor cells (P<0.001). KIF20A overexpression was closely related to poorer overall survival and disease progression-free survival. Furthermore, Cox regression analysis revealed that KIF20A can act as an independent hazard indicator for predicting clinical outcomes in EOC patients. Interestingly, KIF20A overexpression promoted invasion and metastasis of EOC cells and also confers resistance to cisplatin. Conclusion Our findings indicated that KIF20A overexpression predicts unfavorable clinical outcome, revealing that KIF20A holds a promising potential to serve as a useful prognostic biomarker for EOC patients.
Collapse
Affiliation(s)
- Han Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China,
| | - Weijing Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China,
| | - Xiaoying Sun
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China,
| | - Jueming Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China,
| | - Yue Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China,
| | - Chunhao Niu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, Guangdong, China,
| | - Benke Xu
- Department of Anatomy, Medical School of Yangtze University, Jingzhou, China,
| | - Yanna Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China,
| |
Collapse
|
40
|
KIF20A, highly expressed in immature hematopoietic cells, supports the growth of HL60 cell line. Int J Hematol 2018; 108:607-614. [PMID: 30182171 DOI: 10.1007/s12185-018-2527-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/31/2018] [Accepted: 09/03/2018] [Indexed: 10/28/2022]
Abstract
A microtubule-associated motor protein, kinesin-like family member 20A (KIF20A; also called MKlp2) is required for cytokinesis and contributes to intracellular vesicular trafficking. KIF20A plays a critical role in the development of several cancers, but its role in blood cells and hematological malignancies have not been studied. In the present study, we focused on the role of KIF20A in hematopoietic cells and possible involvement in myeloid neoplasms. We found that human leukemia cell lines and normal bone marrow CD34-positive cells stimulated by growth factors, but not mature peripheral blood cells, exhibit high KIF20A expression. We further found that HL60 cells, which originally express a large amount of KIF20A, showed decreased KIF20A expression in parallel with both neutrophil-like and macrophage-like differentiation-induction. KIF20A-knockdown using a lentivirus shRNA transfection system led to partial cell cycle arrest at the G2/M phase and frequent appearance of multinucleated cells. Treatment with a KIF20A-selective inhibitor, paprotrain enhanced the multinuclearity of KIF20A-knockdown cell clones and suppressed growth. The present study contributes to our understanding of the role of KIF20A in blood cells and leukemia cells in particular.
Collapse
|
41
|
Kawai Y, Shibata K, Sakata J, Suzuki S, Utsumi F, Niimi K, Sekiya R, Senga T, Kikkawa F, Kajiyama H. KIF20A expression as a prognostic indicator and its possible involvement in the proliferation of ovarian clear‑cell carcinoma cells. Oncol Rep 2018; 40:195-205. [PMID: 29749467 PMCID: PMC6059742 DOI: 10.3892/or.2018.6401] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 03/16/2018] [Indexed: 11/06/2022] Open
Abstract
Kinesin family member 20A (KIF20A), which is involved in cytokinesis and intracellular transportation, has been recently reported to be upregulated in several malignancies and may contribute to chemotherapeutic resistance. We examined the distribution and expression of KIF20A in clear‑cell carcinoma (CCC) of the ovary to elucidate its clinical significance and molecular mechanism. Paraffin sections from ovarian CCC tissues (N=43) were immunostained with KIF20A antibody, and the staining intensities were semi‑quantitatively evaluated. Furthermore, we investigated whether silencing of KIF20A contributes to the proliferation‑inhibitory potential using CCC cells. During the observational period, 18 patients (41.9%) developed recurrence. The median time to recurrence was 11.5 months. Patients in the high KIF20A expression group showed poorer progression‑free survival (PFS) and overall survival (OS) than those in the low expression group (P=0.0443 and P=0.0478, respectively). In multivariable analyses, KIF20A expression was also a significantly independent indicator of PFS and a marginally significant indicator of OS [PFS: HR (high vs. low), 5.488; 95% CI, 1.410‑24.772 (P=0.0136); OS: HR, 2.835; 95% CI, 0.854‑11.035, (P=0.0897)]. In in vitro studies, the ovarian CCC cell proliferation was significantly decreased by KIF20A silencing or in the presence of KIF20A inhibitor in CCC cells. Cell cycle G2/M arrest and a higher apoptosis‑induced fraction were more frequently observed in si‑KIF20A‑transfected CCC cells than in the control cells. Although the present study was preliminary, these data indicate the possible involvement of KIF20A in the proliferation of CCC, suggesting that targeting this molecule may contribute to reversing the malignant potential consequently affecting the oncologic outcome of CCC patients.
Collapse
Affiliation(s)
- Yosuke Kawai
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| | - Kiyosumi Shibata
- Department of Obstetrics and Gynecology, Banbuntane Hotokukai Hospital, Fujita Health University, Nagoya 454‑8509, Japan
| | - Jun Sakata
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| | - Shiro Suzuki
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| | - Fumi Utsumi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| | - Kaoru Niimi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| | - Ryuichiro Sekiya
- Department of Obstetrics and Gynecology, Banbuntane Hotokukai Hospital, Fujita Health University, Nagoya 454‑8509, Japan
| | - Takeshi Senga
- Department of Internal Medicine, Yahagigawa Hospital, Aichi 444‑1164, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Nagoya University, Nagoya 466‑8550, Japan
| |
Collapse
|
42
|
Xiu G, Sui X, Wang Y, Zhang Z. FOXM1 regulates radiosensitivity of lung cancer cell partly by upregulating KIF20A. Eur J Pharmacol 2018; 833:79-85. [PMID: 29704495 DOI: 10.1016/j.ejphar.2018.04.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/25/2022]
Abstract
Forkhead box protein M1 (FOXM1), an important regulator of tumorigenesis in various human tumors, has recently been reported to play a role in the modulation of radiosensitivity in glioma and breast cancer cells. The present study aimed to investigate the effects of FOXM1 on radiotherapy resistance in human lung cancer and to explore the related molecular mechanisms. The results revealed that FOXM1 expression was upregulated in A549 and H1299 cells after IR (Ionizing radiation). FOXM1 inhibition impeded survival fractions, impeded proliferation, and triggered apoptosis after IR. Moreover, the silencing of FOXM1 dampened cell migration, invasion, and EMT (epithelial-mesenchyman transition) in A549 and H1299 cells treated by IR. In addition, KIF20A was also highly expressed in IR-treated A549 cells and downregulated by FOXM1 inhibition. Knockdown of KIF20A inhibited the survival fraction. Reintroduction of KIF20A partly reversed the effects of FOXM1 on the proliferation, apoptosis, and metastasis of A549 cells. Taken together, these results indicated that FOXM1 might enhance radioresistance partly through the induction of KIF20A expression.
Collapse
Affiliation(s)
- Guanghong Xiu
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China.
| | - Xiujie Sui
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China
| | - Yirong Wang
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China
| | - Ze Zhang
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China
| |
Collapse
|
43
|
Lu M, Huang X, Chen Y, Fu Y, Xu C, Xiang W, Li C, Zhang S, Yu C. Aberrant KIF20A expression might independently predict poor overall survival and recurrence-free survival of hepatocellular carcinoma. IUBMB Life 2018; 70:328-335. [PMID: 29500859 DOI: 10.1002/iub.1726] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 02/07/2018] [Indexed: 12/20/2022]
Abstract
Kinesin family member 20A (KIF20A) is an essential regulator of cytokinesis. In this study, by performing a retrospective study based on data from the Cancer Genome Atlas (TCGA)-Liver and Hepatocellular Carcinoma (LIHC) cohort, we tried to assess the independent prognostic value of KIF20A in terms of overall survival (OS) and recurrence-free survival (RFS). Results showed that normal liver tissues had very low KIF20A expression compared with normal tissues in other cohorts in TCGA. However, the primary HCC tissues (N = 371) had significantly elevated KIF20A expression than normal liver tissues (N = 50). Immunohistochemistry (IHC) data showed that normal hepatocytes had weak KIF20A staining. In comparison, some HCC tissues had medium and strong KIF20A expression, with nuclear-enhanced staining. By grouping patients with primary HCC (N = 365) into high and low KIF20A expression groups, we found that the high expression group had a substantially higher proportion of high-grade tumors (G3/G4) (34/65, 52.3% vs. 96/295, 32.5%, P = 0.0027), advanced tumors (stage III/IV) (28/61, 45.9% vs. 59/280, 21.1%, P < 0.0001) and death (44/67, 65.7% vs. 86/298, 28.9%, P < 0.0001) compared with the low expression group. Kaplan-Meier curves of OS and RFS indicated that high KIF20A expression was associated with worse survival outcomes. Subgroup analysis confirmed the associations in G1/G2, G3/G4 tumors and in early and advanced stages. Following univariate and multivariate analysis revealed that KIF20A expression was an independent prognostic indicator for poor OS (HR: 1.304, 95%CI: 1.157-1.469, P < 0.001) and RFS (HR: 1.144, 95%CI: 1.028-1.272, P < 0.001). Based on these findings, we infer that KIF20A was aberrantly expressed in HCC tissues and its expression might independently predict poor OS and RFS. © 2018 IUBMB Life, 70(4):328-335, 2018.
Collapse
Affiliation(s)
- Mingqin Lu
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoying Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongping Chen
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangyang Fu
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chaona Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Xiang
- Department of Interventional Therapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Li
- Department of Interventional Therapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengguo Zhang
- Department of Infection and Liver Diseases, Liver Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chang Yu
- Department of Interventional Therapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
44
|
Shi C, Huang D, Lu N, Chen D, Zhang M, Yan Y, Deng L, Lu Q, Lu H, Luo S. Aberrantly activated Gli2-KIF20A axis is crucial for growth of hepatocellular carcinoma and predicts poor prognosis. Oncotarget 2018; 7:26206-19. [PMID: 27036048 PMCID: PMC5041975 DOI: 10.18632/oncotarget.8441] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/14/2016] [Indexed: 01/16/2023] Open
Abstract
Glioma-associated oncogene 2 (Gli2), a primary transcriptional regulator of Hedgehog (Hh) signaling, is essential for hepatocellular carcinoma (HCC) growth and survival. However, the underlying molecular mechanism and crucial downstream targets of Gli2 in human HCC are not fully understood. Here, we report the identification of kinesin family member 20A (KIF20A) as a novel downstream target of Gli2, which is important for HCC proliferation and tumor growth. Inhibition of Hh signaling leads to a remarkable decrease of KIF20A expression in HCC cells, whereas overexpression of Gli2 elevates KIF20A expression by activating Forkhead Box M1 (FoxM1)-MMB complex-mediated transcription of this kinesin gene. Gli2-induced HCC cell growth requires enhanced expression of KIF20A, and knockdown of Gli2 or KIF20A represses the proliferation of HCC cells in vitro and in vivo. Correlated with these results, analyses of clinical HCC samples show that Gli2, FoxM1 and KIF20A are highly elevated in primary HCC samples and represent significant risk factors for HCC recurrence and survival. Conclusion: KIF20A is an important downstream target gene of Hh signaling. And, the Gli2-KIF20A axis is essential for the proliferation and growth of human HCC cells. Our study also suggests Gli2-KIF20A axis as a potential target for future therapeutic intervention and as an independent prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Chao Shi
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dengliang Huang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dan Chen
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Minhong Zhang
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yehong Yan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Libin Deng
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, China
| | - Quqin Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Hua Lu
- Department of Biochemistry and Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
45
|
Buettner F, Pratanwanich N, McCarthy DJ, Marioni JC, Stegle O. f-scLVM: scalable and versatile factor analysis for single-cell RNA-seq. Genome Biol 2017; 18:212. [PMID: 29115968 PMCID: PMC5674756 DOI: 10.1186/s13059-017-1334-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/29/2017] [Indexed: 11/10/2022] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) allows studying heterogeneity in gene expression in large cell populations. Such heterogeneity can arise due to technical or biological factors, making decomposing sources of variation difficult. We here describe f-scLVM (factorial single-cell latent variable model), a method based on factor analysis that uses pathway annotations to guide the inference of interpretable factors underpinning the heterogeneity. Our model jointly estimates the relevance of individual factors, refines gene set annotations, and infers factors without annotation. In applications to multiple scRNA-seq datasets, we find that f-scLVM robustly decomposes scRNA-seq datasets into interpretable components, thereby facilitating the identification of novel subpopulations.
Collapse
Affiliation(s)
- Florian Buettner
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
- Current address: Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.
| | - Naruemon Pratanwanich
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Davis J McCarthy
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
- St Vincent's Institute of Medical Research, 41 Victoria Parade, Fitzroy, Victoria, 3065, Australia
| | - John C Marioni
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
- Cancer Research UK Cambridge Institute, Cambridge, UK.
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Meyerhofstr. 1, 69117, Heidelberg, Germany.
| |
Collapse
|
46
|
Atherton J, Yu IM, Cook A, Muretta JM, Joseph A, Major J, Sourigues Y, Clause J, Topf M, Rosenfeld SS, Houdusse A, Moores CA. The divergent mitotic kinesin MKLP2 exhibits atypical structure and mechanochemistry. eLife 2017; 6:27793. [PMID: 28826477 PMCID: PMC5602324 DOI: 10.7554/elife.27793] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Accepted: 08/07/2017] [Indexed: 01/17/2023] Open
Abstract
MKLP2, a kinesin-6, has critical roles during the metaphase-anaphase transition and cytokinesis. Its motor domain contains conserved nucleotide binding motifs, but is divergent in sequence (~35% identity) and size (~40% larger) compared to other kinesins. Using cryo-electron microscopy and biophysical assays, we have undertaken a mechanochemical dissection of the microtubule-bound MKLP2 motor domain during its ATPase cycle, and show that many facets of its mechanism are distinct from other kinesins. While the MKLP2 neck-linker is directed towards the microtubule plus-end in an ATP-like state, it does not fully dock along the motor domain. Furthermore, the footprint of the MKLP2 motor domain on the MT surface is altered compared to motile kinesins, and enhanced by kinesin-6-specific sequences. The conformation of the highly extended loop6 insertion characteristic of kinesin-6s is nucleotide-independent and does not contact the MT surface. Our results emphasize the role of family-specific insertions in modulating kinesin motor function. Cells constantly replicate to provide new cells for growing tissues, and to replace ageing or defective cells around the body. Each new cell needs a copy of the genetic material, and a cellular structure called the mitotic spindle makes sure that this material is shared correctly when a cell divides in two. The spindle is built from protein filaments called microtubules, and the protein filaments grow and shrink as the mitotic spindle carries out its role. Many of these changes in the spindle are driven by proteins called molecular motors, which break down energy-rich molecules of ATP to power them as they walk along the filaments. Kinesins, for example, are molecular motors that can move along microtubules and there are over 40 different kinesins encoded in the human genome. More than half of the human kinesins are involved in cell division including one called MKLP2. Little is known about MKLP2 but some earlier findings had suggested that it would behave very differently compared to other kinesins. Understanding how a kinesin motor works requires studying it in complex with its microtubule tracks. Atherton, Yu et al. have now used a technique called cryo-electron microscopy – which is uniquely suited to looking at large and complicated samples in three dimensions – to observe how the motor in MKLP2 changes shape as it works. This revealed that, while MKLP2 works in a fundamentally similar way to other kinesins, many aspects of its molecular mechanism are highly unusual. These include how it binds to the microtubule, how it interacts with ATP and how it generates force. These findings show that there is much greater diversity in the molecular mechanisms of the kinesins involved in cell division than was previously thought. Several anticancer drugs target kinesins to stop cells dividing and so this diversity may make it easier to target only certain kinesins with drugs, which in turn would have fewer side effects. First, though, it will be important to find out how the unusual mechanism of MKLP2 coordinates and influences other components of the spindle to reveal a fuller picture of what happens when cells replicate.
Collapse
Affiliation(s)
- Joseph Atherton
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
| | - I-Mei Yu
- Structural Motility, Institut Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Paris, France
| | - Alexander Cook
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
| | - Joseph M Muretta
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, United Sates
| | - Agnel Joseph
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
| | - Jennifer Major
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Yannick Sourigues
- Structural Motility, Institut Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Paris, France
| | - Jeffrey Clause
- Structural Motility, Institut Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Paris, France
| | - Maya Topf
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
| | - Steven S Rosenfeld
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Anne Houdusse
- Structural Motility, Institut Curie, Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Paris, France
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Birkbeck College, London, United Kingdom
| |
Collapse
|
47
|
Weiler SME, Pinna F, Wolf T, Lutz T, Geldiyev A, Sticht C, Knaub M, Thomann S, Bissinger M, Wan S, Rössler S, Becker D, Gretz N, Lang H, Bergmann F, Ustiyan V, Kalin TV, Singer S, Lee JS, Marquardt JU, Schirmacher P, Kalinichenko VV, Breuhahn K. Induction of Chromosome Instability by Activation of Yes-Associated Protein and Forkhead Box M1 in Liver Cancer. Gastroenterology 2017; 152:2037-2051.e22. [PMID: 28249813 DOI: 10.1053/j.gastro.2017.02.018] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/07/2017] [Accepted: 02/19/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Many different types of cancer cells have chromosome instability. The hippo pathway leads to phosphorylation of the transcriptional activator yes-associated protein 1 (YAP1, YAP), which regulates proliferation and has been associated with the development of liver cancer. We investigated the effects of hippo signaling via YAP on chromosome stability and hepatocarcinogenesis in humans and mice. METHODS We analyzed transcriptome data from 242 patients with hepatocellular carcinoma (HCC) to search for gene signatures associated with chromosomal instability (CIN); we investigated associations with overall survival time and cancer recurrence using Kaplan-Meier curves. We analyzed changes in expression of these signature genes, at mRNA and protein levels, after small interfering RNA-mediated silencing of YAP in Sk-Hep1, SNU182, HepG2, or pancreatic cancer cells, as well as incubation with thiostrepton (an inhibitor of forkhead box M1 [FOXM1]) or verteporfin (inhibitor of the interaction between YAP and TEA domain transcription factor 4 [TEAD4]). We performed co-immunoprecipitation and chromatin immunoprecipitation experiments. We collected liver tissues from mice that express a constitutively active form of YAP (YAPS127A) and analyzed gene expression signatures and histomorphologic parameters associated with chromosomal instability. Mice were given injections of thiostrepton and livers were collected and analyzed by immunoblotting, immunohistochemistry, histology, and real-time polymerase chain reaction. We performed immunohistochemical analyses on tissue microarrays of 105 HCCs and 7 nontumor liver tissues. RESULTS Gene expression patterns associated with chromosome instability, called CIN25 and CIN70, were detected in HCCs from patients with shorter survival time or early cancer recurrence. TEAD4 and YAP were required for CIN25 and CIN70 signature expression via induction and binding of FOXM1. Disrupting the interaction between YAP and TEAD4 with verteporfin, or inhibiting FOXM1 with thiostrepton, reduced the chromosome instability gene expression patterns. Hyperplastic livers and tumors from YAPS127A mice had increased CIN25 and CIN70 gene expression patterns, aneuploidy, and defects in mitosis. Injection of YAPS127A mice with thiostrepton reduced liver overgrowth and signs of chromosomal instability. In human HCC tissues, high levels of nuclear YAP correlated with increased chromosome instability gene expression patterns and aneuploidy. CONCLUSIONS By analyzing cell lines, genetically modified mice, and HCC tissues, we found that YAP cooperates with FOXM1 to contribute to chromosome instability. Agents that disrupt this pathway might be developed as treatments for liver cancer. Transcriptome data are available in the Gene Expression Omnibus public database (accession numbers: GSE32597 and GSE73396).
Collapse
Affiliation(s)
- Sofia M E Weiler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Federico Pinna
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Wolf
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Teresa Lutz
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Aman Geldiyev
- International Educational-Scientific Center, Ashgabat City, Turkmenistan
| | - Carsten Sticht
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Maria Knaub
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Thomann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michaela Bissinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Shan Wan
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephanie Rössler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Diana Becker
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Hauke Lang
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Vladimir Ustiyan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tatiana V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stephan Singer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jens U Marquardt
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Vladimir V Kalinichenko
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
48
|
Saito K, Ohta S, Kawakami Y, Yoshida K, Toda M. Functional analysis of KIF20A, a potential immunotherapeutic target for glioma. J Neurooncol 2017; 132:63-74. [DOI: 10.1007/s11060-016-2360-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 12/23/2016] [Indexed: 01/05/2023]
|
49
|
High Expression of KIF20A Is Associated with Poor Overall Survival and Tumor Progression in Early-Stage Cervical Squamous Cell Carcinoma. PLoS One 2016; 11:e0167449. [PMID: 27941992 PMCID: PMC5152822 DOI: 10.1371/journal.pone.0167449] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/14/2016] [Indexed: 12/05/2022] Open
Abstract
Background The kinesin family member 20a (KIF20A) protein has been implicated in the development and progression of many human cancers; however, its precise function and role in cervical cancer remain largely unclear. This study aimed to investigate the expression profile and prognostic value of KIF20A in patients with early-stage cervical squamous cell carcinoma. Methods We examined the mRNA and protein levels of KIF20A in eight cervical cancer cell lines and eight paired cervical cancer samples, compared with normal cervical epithelial cells and adjacent normal cervical tissues, respectively. Immunohistochemistry was performed to detect the expression of KIF20A in paraffin-embedded specimens from 169 early-stage cervical squamous cell carcinoma patients. Statistical analyses were applied to analyze the association between KIF20A expression and clinical variables, as well with patient survival. Results The mRNA and protein expression levels of KIF20A were significantly elevated in cervical cancer cell lines and lesions compared with normal cells and corresponding normal tissues (P < 0.05). Immunohistochemistry analysis in 169 cervical cancer cases revealed that increased KIF20A expression was strongly associated with human papillomavirus (HPV) infection (P = 0.008), clinical stage (P = 0.001), tumor recurrence (P = 0.016), vital status (P < 0.001), the property of the surgical margin (P = 0.032), the lymphovascular space involvement (P = 0.014), and pelvic lymph node metastasis (P = 0.001). The overall survival and disease-free survival of patients with high levels of KIF20A expression were significantly poorer than those with low KIF20A expression. KIF20A was an independent survival prognostic factor, as evidenced by univariate and multivariate analysis. Conclusions Our results illustrate that elevated KIF20A expression correlates with HPV infection, clinical stage, tumor recurrence, lymphovascular space involvement, pelvic lymph node metastasis, and poor outcome in early-stage cervical squamous cell carcinoma patients. KIF20A aberrant expression is a novel independent unfavorable prognostic factor and may present a potential therapeutic target for cervical cancer.
Collapse
|
50
|
De Iuliis F, Taglieri L, Salerno G, Giuffrida A, Milana B, Giantulli S, Carradori S, Silvestri I, Scarpa S. The kinesin Eg5 inhibitor K858 induces apoptosis but also survivin-related chemoresistance in breast cancer cells. Invest New Drugs 2016; 34:399-406. [PMID: 26994617 DOI: 10.1007/s10637-016-0345-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/14/2016] [Indexed: 01/29/2023]
Abstract
Inhibitors of kinesin spindle protein Eg5 are characterized by pronounced antitumor activity. Our group has recently synthesized and screened a library of 1,3,4-thiadiazoline analogues with the pharmacophoric structure of K858, an Eg5 inhibitor. We herein report the effects of K858 on four different breast cancer cell lines: MCF7 (luminal A), BT474 (luminal B), SKBR3 (HER2 like) and MDA-MB231 (basal like). We demonstrated that K858 displayed anti-proliferative activity on every analyzed breast cancer cell line by inducing apoptosis. However, at the same time, we showed that K858 up-regulated survivin, an anti-apoptotic molecule. We then performed a negative regulation of survivin expression, with the utilization of wortmannin, an AKT inhibitor, and obtained a significant increase of K858-dependent apoptosis. These data demonstrate that K858 is a potent inhibitor of replication and induces apoptosis in breast tumor cells, independently from the tumor phenotype. This anti-proliferative response of tumor cells to K858 can be limited by the contemporaneous over-expression of survivin; consequently, the reduction of survivin levels, obtained with AKT inhibitors, can sensitize tumor cells to K858-induced apoptosis.
Collapse
Affiliation(s)
- Francesca De Iuliis
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Ludovica Taglieri
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Gerardo Salerno
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Anna Giuffrida
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Bernardina Milana
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Sabrina Giantulli
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Simone Carradori
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, 66100, Chieti, Italy
| | - Ida Silvestri
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Susanna Scarpa
- Department of Experimental Medicine, Sapienza University, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|