1
|
Liang X, Li L, Fan Y. Diagnostic, Prognostic, and Immunological Roles of HELLS in Pan-Cancer: A Bioinformatics Analysis. Front Immunol 2022; 13:870726. [PMID: 35774795 PMCID: PMC9237247 DOI: 10.3389/fimmu.2022.870726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Background Inappropriate repair of DNA damage drives carcinogenesis. Lymphoid-specific helicase (HELLS) is an important component of the chromatin remodeling complex that helps repair DNA through various mechanisms such as DNA methylation, histone posttranslational modification, and nucleosome remodeling. Its role in human cancer initiation and progression has garnered recent attention. Our study aims to provide a more systematic and comprehensive understanding of the role of HELLS in the development and progression of multiple malignancies through analysis of HELLS in cancers. Methods We explored the role of HELLS in cancers using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) database. Multiple web platforms and software were used for data analysis, including R, Cytoscape, HPA, Archs4, TISIDB, cBioPortal, STRING, GSCALite, and CancerSEA. Results High HELLS expression was found in a variety of cancers and differentially expressed across molecular and immune subtypes. HELLS was involved in many cancer pathways. Its expression positively correlated with Th2 and Tcm cells in most cancers. It also correlated with genetic markers of immunomodulators in various cancers. Conclusions Our study elucidates the role HELLS plays in promotion, inhibition, and treatment of different cancers. HELLS is a potential cancer diagnostic and prognostic biomarker with immune, targeted, or cytotoxic therapeutic value. This work is a prerequisite to clinical validation and treatment of HELLS in cancers.
Collapse
Affiliation(s)
- Xiao Liang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Linji Li
- Department of Anesthesiology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Yuchao Fan
- Department of Anesthesiology, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Yuchao Fan,
| |
Collapse
|
2
|
CEP55 Positively Affects Tumorigenesis of Esophageal Squamous Cell Carcinoma and Is Correlated with Poor Prognosis. JOURNAL OF ONCOLOGY 2021; 2021:8890715. [PMID: 34104194 PMCID: PMC8159646 DOI: 10.1155/2021/8890715] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/16/2021] [Accepted: 03/27/2021] [Indexed: 01/19/2023]
Abstract
Centrosomal protein 55 (CEP55) is a centrosome- and midbody-associated protein that is overexpressed in several cancers. However, the underlying molecular mechanism of CEP55-mediated progression and metastasis of esophageal squamous cell carcinoma (ESCC) is not clear. In the current study, we detected CEP55 mRNA by qRT-PCR while protein expression was detected by western blot analysis and immunohistochemistry (IHC). In addition, we knocked down CEP55 and investigated the ability of CEP55 to affect colony formation and migration. Here, we report that CEP55 mRNA and protein expression was significantly increased in ESCC. IHC staining showed that CEP55 expression correlated with TNM stage (p=0.046) and lymph node metastases (p=0.024). According to overall survival (OS) and disease-free survival (DFS), patients whose tumors expressed a higher level of CEP55 had a poorer prognosis than those with low expression level of CEP55. A multivariate analysis revealed that CEP55 expression was an independent prognostic indicator for patients with ESCC. Knockdown of CEP55 decreased the colony formation ability and migration of ESCC cells and also reduced the phosphorylation of Src, FAK, and ERK. Therefore, our study implied that CEP55 may be a valuable biomarker and a potential target in the treatment of patients with ESCC.
Collapse
|
3
|
Huang D, Li Q, Sun X, Sun X, Tang Y, Qu Y, Liu D, Yu T, Li G, Tong T, Zhang Y. CRL4 DCAF8 dependent opposing stability control over the chromatin remodeler LSH orchestrates epigenetic dynamics in ferroptosis. Cell Death Differ 2021; 28:1593-1609. [PMID: 33288900 PMCID: PMC8166945 DOI: 10.1038/s41418-020-00689-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/06/2020] [Accepted: 11/16/2020] [Indexed: 01/28/2023] Open
Abstract
Despite the emerging evidence on ferroptosis implicated in diverse pathologies, molecular linkage between oxidative inducers and chromatin as epigenetic memory carrier for its propagation remains elusive. Here, we report the identification of two WD40 proteins DCAF8 and WDR76 as substrate adapter and molecular inhibitor respectively of the Cullin-4 RING ubiquitin ligase (CRL4) system for stability control of chromatin remodeler LSH. Degradation analysis and CRL4-DCAF8 complex reconstitution demonstrate that CRL4DCAF8 is a bona fide E3 ligase for LSH. In contrast, WDR76 antagonizes DCAF8-targeted LSH proteolysis through competitive inhibition of the holo-CRL4DCAF8-LSH complex assembly. Importantly, this opposing regulatory strategy is utilized in lipid hydroperoxide induced ferroptosis, where we identify key redox homeostasis genes significantly regulated by the DCAF8/WDR76/LSH axis through transcriptomic epistasis analysis. This regulation is mechanistically attributed to DNA hydroxymethylation fostered WDR76 interaction with LSH and increased ratio of DCAF8 to WDR76 for antagonistic LSH association accompanying decreased DNA oxidation along with ROS overproduction. Evaluation of epigenetic dynamics at ferroptosis gene promoters reveals linker histone H1- and LSH-associated transcriptional repression is coordinately removed upon lipid peroxidation stress. Together with the phenotypes driven by WDR76 and DCAF8 manipulations, these data identify DCAF8- and WDR76-adapted oxidative damage sensing through DNA hydroxymethylation for LSH degradation control as a crucial nexus in epigenetic regulation of ferroptosis.
Collapse
Affiliation(s)
- Daoyuan Huang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081, Beijing, China
| | - Xinpei Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Xiwen Sun
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Yunyi Tang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Yanan Qu
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Dawei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081, Beijing, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, 100081, Beijing, China
| | - Guodong Li
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Tanjun Tong
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China
| | - Yu Zhang
- Peking University Research Center on Aging, Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 100191, Beijing, China.
| |
Collapse
|
4
|
Tabatabaei Dakhili SA, Pérez DJ, Gopal K, Haque M, Ussher JR, Kashfi K, Velázquez-Martínez CA. SP1-independent inhibition of FOXM1 by modified thiazolidinediones. Eur J Med Chem 2020; 209:112902. [PMID: 33069434 DOI: 10.1016/j.ejmech.2020.112902] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 11/25/2022]
Abstract
This research article describes an approach to modify the thiazolidinedione scaffold to produce test drugs capable of binding to, and inhibit, the in vitro transcriptional activity of the oncogenic protein FOXM1. This approach allowed us to obtain FOXM1 inhibitors that bind directly to the FOXM1-DNA binding domain without targeting the expression levels of Sp1, an upstream transcription factor protein known to activate the expression of FOXM1. Briefly, we modified the chemical structure of the thiazolidinedione scaffold present in anti-diabetic medications such as pioglitazone, rosiglitazone and the former anti-diabetic drug troglitazone, because these drugs have been reported to exert inhibition of FOXM1 but hit other targets as well. After the chemical synthesis of 11 derivatives possessing a modified thiazolidinedione moiety, we screened all test compounds using in vitro protocols to measure their ability to (a) dissociate a FOXM1-DNA complex (EMSA assay); (b) decrease the expression of FOXM1 in triple negative-breast cancer cells (WB assay); (c) downregulate the expression of FOXM1 downstream targets (luciferase reporter assays and qPCR); and inhibit the formation of colonies of MDA-MB-231 cancer cells (colony formation assay). We also identified a potential binding mode associated with these compounds in which compound TFI-10, one of the most active molecules, exerts binding interactions with Arg289, Trp308, and His287. Unlike the parent drug, troglitazone, compound TFI-10 does not target the in vitro expression of Sp1, suggesting that it is possible to design FOXM1 inhibitors with a better selectivity profile.
Collapse
Affiliation(s)
| | - David J Pérez
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Unidad Radiofarmacia-Ciclotrón, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Moinul Haque
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Khosrow Kashfi
- Department of Molecular, Cellular, & Biomedical Sciences, City University of New York School of Medicine, New York, USA; Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | | |
Collapse
|
5
|
Nie S, Lou L, Wang J, Cui J, Wu W, Zhang Q, Liu Y, Su L, Chang Y, Guo W, Shen H, Xing L, Li Y. Expression, association with clinicopathological features and prognostic potential of CEP55, p-Akt, FoxM1 and MMP-2 in astrocytoma. Oncol Lett 2020; 20:1685-1694. [PMID: 32724411 PMCID: PMC7377175 DOI: 10.3892/ol.2020.11742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 04/27/2020] [Indexed: 11/19/2022] Open
Abstract
Centrosomal protein 55 (CEP55) is a member of the centrosomal-associated protein family and participates in the regulation of cytokinesis during cell mitosis. However, aberrant CEP55 protein expression has been observed in human tumors. In addition, CEP55 regulates the biological functions of tumors by inducing the Akt pathway and upregulating forkhead box protein M1 (FoxM1) and matrix metalloproteinase-2 (MMP-2). In the present study, the levels, clinicopathological features and prognostic potential of CEP55, phosphorylated Akt (p-Akt), FoxM1 and MMP-2 in astrocytoma were evaluated. CEP55, p-Akt, FoxM1 and MMP-2 levels were examined in 27 normal brain tissues and 262 astrocytoma tissues by using immunohistochemistry. Furthermore, Kaplan-Meier analysis and Cox proportional hazards models were applied to predict the prognosis of patients with astrocytoma. The results indicated that expression levels of CEP55 and other proteins were elevated in human astrocytoma compared with those in normal brain tissue. The levels of the selected proteins were increased as the tumor grade increased. Furthermore, CEP55 expression was positively correlated with p-Akt, FoxM1 and MMP-2 levels in astrocytoma. Overall survival analysis revealed that patient prognosis was associated with CEP55, p-Akt, FoxM1 and MMP-2 levels, as well as with the tumor grade and patient age. Furthermore, CEP55, FoxM1, tumor grade and patient age were independent prognostic factors in astrocytoma according to multivariate analysis. Taken together, the present results suggested that CEP55, p-Akt, FoxM1 and MMP-2 have crucial roles in the progression and prognosis of human astrocytoma and that CEP55 and FoxM1 may be potential therapeutic targets.
Collapse
Affiliation(s)
- Saisai Nie
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Lei Lou
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Juan Wang
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jinfeng Cui
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Wenxin Wu
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Qing Zhang
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Ying Liu
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Lingrui Su
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Ying Chang
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Wenli Guo
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lingxiao Xing
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yuehong Li
- Department of Pathology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
6
|
Kollárovič G, Topping CE, Shaw EP, Chambers AL. The human HELLS chromatin remodelling protein promotes end resection to facilitate homologous recombination and contributes to DSB repair within heterochromatin. Nucleic Acids Res 2020; 48:1872-1885. [PMID: 31802118 PMCID: PMC7038987 DOI: 10.1093/nar/gkz1146] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 11/23/2022] Open
Abstract
Efficient double-strand break repair in eukaryotes requires manipulation of chromatin structure. ATP-dependent chromatin remodelling enzymes facilitate different DNA repair pathways, during different stages of the cell cycle and in varied chromatin environments. The contribution of remodelling factors to double-strand break repair within heterochromatin during G2 is unclear. The human HELLS protein is a Snf2-like chromatin remodeller family member and is mutated or misregulated in several cancers and some cases of ICF syndrome. HELLS has been implicated in the DNA damage response, but its mechanistic function in repair is not well understood. We discover that HELLS facilitates homologous recombination at two-ended breaks and contributes to repair within heterochromatic regions during G2. HELLS promotes initiation of HR by facilitating end-resection and accumulation of CtIP at IR-induced foci. We identify an interaction between HELLS and CtIP and establish that the ATPase domain of HELLS is required to promote DSB repair. This function of HELLS in maintenance of genome stability is likely to contribute to its role in cancer biology and demonstrates that different chromatin remodelling activities are required for efficient repair in specific genomic contexts.
Collapse
Affiliation(s)
- Gabriel Kollárovič
- DNA-protein Interactions Unit, School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Caitríona E Topping
- DNA-protein Interactions Unit, School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Edward P Shaw
- DNA-protein Interactions Unit, School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Anna L Chambers
- DNA-protein Interactions Unit, School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
7
|
Seyedabadi S, Saidijam M, Najafi R, Mousavi-Bahar SH, Jafari M, MohammadGanji S, Mahdavinezhad A. Assessment of CEP55, PLK1 and FOXM1 expression in patients with bladder cancer in comparison with healthy individuals. Cancer Invest 2018; 36:407-414. [PMID: 30277841 DOI: 10.1080/07357907.2018.1514504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/18/2018] [Indexed: 01/04/2023]
Abstract
This case/control study is aimed at investigating the expression of CEP55, PLK1 and FOXM1 in bladder cancer tissues and comparing it with healthy tissue and their relationship with clinicopathological features of BC. Total RNA was extracted; then, gene expression was performed using real-time PCR relative to 18 s rRNA. 2-ΔΔCT method was used to calculate the relative expression of genes. A significant over expression of FOXM1, PLK1 and CEP55 was observed in tumor samples compared to adjacent and normal bladder tissues (all p = 0.001). Therefore, they may be supposed as potential candidate's biomarkers for early diagnosis and targets for cancer therapy.
Collapse
Affiliation(s)
- Saman Seyedabadi
- a Research Center for Molecular Medicine and Genetics , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Massoud Saidijam
- a Research Center for Molecular Medicine and Genetics , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Rezvan Najafi
- a Research Center for Molecular Medicine and Genetics , Hamadan University of Medical Sciences , Hamadan , Iran
| | | | - Mohammad Jafari
- c Department of Pathology , Medical School, Hamadan University of Medical Sciences , Hamadan , Iran
| | - Sajjad MohammadGanji
- a Research Center for Molecular Medicine and Genetics , Hamadan University of Medical Sciences , Hamadan , Iran
| | - Ali Mahdavinezhad
- a Research Center for Molecular Medicine and Genetics , Hamadan University of Medical Sciences , Hamadan , Iran
| |
Collapse
|
8
|
Kalimutho M, Sinha D, Jeffery J, Nones K, Srihari S, Fernando WC, Duijf PH, Vennin C, Raninga P, Nanayakkara D, Mittal D, Saunus JM, Lakhani SR, López JA, Spring KJ, Timpson P, Gabrielli B, Waddell N, Khanna KK. CEP55 is a determinant of cell fate during perturbed mitosis in breast cancer. EMBO Mol Med 2018; 10:e8566. [PMID: 30108112 PMCID: PMC6127888 DOI: 10.15252/emmm.201708566] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 07/15/2018] [Accepted: 07/18/2018] [Indexed: 12/28/2022] Open
Abstract
The centrosomal protein, CEP55, is a key regulator of cytokinesis, and its overexpression is linked to genomic instability, a hallmark of cancer. However, the mechanism by which it mediates genomic instability remains elusive. Here, we showed that CEP55 overexpression/knockdown impacts survival of aneuploid cells. Loss of CEP55 sensitizes breast cancer cells to anti-mitotic agents through premature CDK1/cyclin B activation and CDK1 caspase-dependent mitotic cell death. Further, we showed that CEP55 is a downstream effector of the MEK1/2-MYC axis. Blocking MEK1/2-PLK1 signaling therefore reduced outgrowth of basal-like syngeneic and human breast tumors in in vivo models. In conclusion, high CEP55 levels dictate cell fate during perturbed mitosis. Forced mitotic cell death by blocking MEK1/2-PLK1 represents a potential therapeutic strategy for MYC-CEP55-dependent basal-like, triple-negative breast cancers.
Collapse
Affiliation(s)
- Murugan Kalimutho
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Debottam Sinha
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Jessie Jeffery
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Katia Nones
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- Queensland Centre for Medical Genomics, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Qld, Australia
| | - Sriganesh Srihari
- Computational Systems Biology Laboratory, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Qld, Australia
| | | | - Pascal Hg Duijf
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Claire Vennin
- Cancer Division, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Prahlad Raninga
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | | | - Deepak Mittal
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Jodi M Saunus
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia
| | - Sunil R Lakhani
- Centre for Clinical Research, The University of Queensland, Herston, Qld, Australia
- School of Medicine, The University of Queensland, Herston, Qld, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, Qld, Australia
| | - J Alejandro López
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
- School of Natural Sciences, Griffith University, Nathan, Qld, Australia
| | - Kevin J Spring
- Liverpool Clinical School, University of Western Sydney, Liverpool, NSW, Australia
- Ingham Institute, Liverpool Hospital, Liverpool, NSW, Australia
- South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW, Australia
| | - Paul Timpson
- Cancer Division, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, NSW, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Brian Gabrielli
- University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
- Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Qld, Australia
| | - Nicola Waddell
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| |
Collapse
|
9
|
Kabir MF, Mohd Ali J, Haji Hashim O. Microarray gene expression profiling in colorectal (HCT116) and hepatocellular (HepG2) carcinoma cell lines treated with Melicope ptelefolia leaf extract reveals transcriptome profiles exhibiting anticancer activity. PeerJ 2018; 6:e5203. [PMID: 30042885 PMCID: PMC6054789 DOI: 10.7717/peerj.5203] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Abstract
Background We have previously reported anticancer activities of Melicope ptelefolia (MP) leaf extracts on four different cancer cell lines. However, the underlying mechanisms of actions have yet to be deciphered. In the present study, the anticancer activity of MP hexane extract (MP-HX) on colorectal (HCT116) and hepatocellular carcinoma (HepG2) cell lines was characterized through microarray gene expression profiling. Methods HCT116 and HepG2 cells were treated with MP-HX for 24 hr. Total RNA was extracted from the cells and used for transcriptome profiling using Applied Biosystem GeneChip™ Human Gene 2.0 ST Array. Gene expression data was analysed using an Applied Biosystems Expression Console and Transcriptome Analysis Console software. Pathway enrichment analyses was performed using Ingenuity Pathway Analysis (IPA) software. The microarray data was validated by profiling the expression of 17 genes through quantitative reverse transcription PCR (RT-qPCR). Results MP-HX induced differential expression of 1,290 and 1,325 genes in HCT116 and HepG2 cells, respectively (microarray data fold change, MA_FC ≥ ±2.0). The direction of gene expression change for the 17 genes assayed through RT-qPCR agree with the microarray data. In both cell lines, MP-HX modulated the expression of many genes in directions that support antiproliferative activity. IPA software analyses revealed MP-HX modulated canonical pathways, networks and biological processes that are associated with cell cycle, DNA replication, cellular growth and cell proliferation. In both cell lines, upregulation of genes which promote apoptosis, cell cycle arrest and growth inhibition were observed, while genes that are typically overexpressed in diverse human cancers or those that promoted cell cycle progression, DNA replication and cellular proliferation were downregulated. Some of the genes upregulated by MP-HX include pro-apoptotic genes (DDIT3, BBC3, JUN), cell cycle arresting (CDKN1A, CDKN2B), growth arrest/repair (TP53, GADD45A) and metastasis suppression (NDRG1). MP-HX downregulated the expression of genes that could promote anti-apoptotic effect, cell cycle progression, tumor development and progression, which include BIRC5, CCNA2, CCNB1, CCNB2, CCNE2, CDK1/2/6, GINS2, HELLS, MCM2/10 PLK1, RRM2 and SKP2. It is interesting to note that all six top-ranked genes proposed to be cancer-associated (PLK1, MCM2, MCM3, MCM7, MCM10 and SKP2) were downregulated by MP-HX in both cell lines. Discussion The present study showed that the anticancer activities of MP-HX are exerted through its actions on genes regulating apoptosis, cell proliferation, DNA replication and cell cycle progression. These findings further project the potential use of MP as a nutraceutical agent for cancer therapeutics.
Collapse
Affiliation(s)
- Mohammad Faujul Kabir
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Johari Mohd Ali
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Onn Haji Hashim
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
10
|
Liu X, Zhang L, Liu Y, Cui J, Che S, An X, Song Y, Cao B. Circ-8073 regulates CEP55 by sponging miR-449a to promote caprine endometrial epithelial cells proliferation via the PI3K/AKT/mTOR pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1130-1147. [PMID: 29800603 DOI: 10.1016/j.bbamcr.2018.05.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/11/2018] [Accepted: 05/21/2018] [Indexed: 01/26/2023]
Abstract
Circular RNAs (circRNAs) are a large class of endogenous non-coding RNAs that function as regulators in various cells and tissues. Here, the function and mechanism of circRNA8073 (Circ-8073) on endometrial epithelial cells (EECs) and the development of endometrial receptivity were investigated in dairy goats. Circ-8073 could bind to and inhibit miR-449a activity. Circ-8073 binding to the target site of miR-449a had a negative feedback relationship. Centrosomal protein55 (CEP55) was a direct target gene of miR-449a, and Circ-8073 could increase the expression levels of CEP55 by sponging miR-449a in EECs in vitro. Circ-8073/miR-449a/CEP55 could promote EECs proliferation via the PI3K/AKT/mTOR pathway. In addition, CEP55 could regulate the expression levels of vascular endothelial growth factor (VEGF) and forkhead box M1 (FOXM1) in EECs, which contributed to the development of endometrial receptivity. These findings showed that Circ-8073 regulated CEP55 by sponging miR-449a to promote EEC proliferation via the PI3K/AKT/mTOR pathway, suggesting that it could function as a regulator in the development of endometrial receptivity in dairy goats.
Collapse
Affiliation(s)
- Xiaorui Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuexia Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Jiuzeng Cui
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Sicheng Che
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Binyun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
11
|
Hoogland AM, Böttcher R, Verhoef E, Jenster G, van Leenders GJLH. Gene-expression analysis of gleason grade 3 tumor glands embedded in low- and high-risk prostate cancer. Oncotarget 2018; 7:37846-37856. [PMID: 27191985 PMCID: PMC5122354 DOI: 10.18632/oncotarget.9344] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 04/25/2016] [Indexed: 12/02/2022] Open
Abstract
The Gleason score (GS) of prostate cancer on diagnostic biopsies is an important parameter for therapeutic decision-making. Biopsy GS under-estimates the actual GS at radical prostatectomy in a significant number of patients due to sampling artifact. The aim of this study was to identify markers that are differentially expressed in Gleason grade 3 (GG3) tumor glands embedded in GS 4 + 3 = 7 and GS 3 + 3 = 6 prostate cancer using laser capture microdissection and RNA sequencing. GG3 tumor glands embedded in nine GS 3 + 3 = 6 and nine GS 4 + 3 = 7 prostate cancers were isolated by laser capture microdissection of frozen radical prostatectomy specimens. After RNA amplification and RNA sequencing, differentially expressed genes in both GG3 components were identified by a 2log fold change > 1.0 and p-value < 0.05. We applied immunohistochemistry on a tissue micro-array representing 481 radical prostatectomy samples for further validation on protein level. A total of 501 genes were up-regulated and 421 down-regulated in GG3 glands embedded in GS 4 + 3 = 7 as compared to GS 3 + 3 = 6 prostate cancer. We selected HELLS, ZIC2 and ZIC5 genes for further validation. ZIC5 mRNA was up-regulated 17 fold (p = 8.4E–07), ZIC2 8 fold (p = 1.3E–05) and HELLS 2 fold (p = 0.006) in GG3 glands derived from GS 4 + 3 = 7. HELLS expression of ≥ 1% occurred in 10% GS < 7, 17% GS 7 and 43% GS >7 prostate cancer (p < 0.001). Using a cut-off of ≥ 1%, protein expression of ZIC5 was present in 28% GS < 7, 43% GS 7 and 57% GS > 7 cancer (p < 0.001). ZIC2 was neither associated with GS nor outcome in our validation set. HELLS was independently predictive for biochemical-recurrence after radical prostatectomy (HR 2.3; CI 1.5–3.6; p < 0.01). In conclusion, HELLS and ZIC5 might be promising candidate markers for selection of biopsy GS 6 prostate cancer being at risk for up-grading at prostatectomy.
Collapse
Affiliation(s)
- A Marije Hoogland
- Departments of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - René Böttcher
- Departments of Urology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Bioinformatics, University of Applied Sciences Wildau, Wildau, Germany
| | - Esther Verhoef
- Departments of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guido Jenster
- Departments of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
12
|
USP5 promotes tumorigenesis and progression of pancreatic cancer by stabilizing FoxM1 protein. Biochem Biophys Res Commun 2017; 492:48-54. [PMID: 28807830 DOI: 10.1016/j.bbrc.2017.08.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 08/10/2017] [Indexed: 02/08/2023]
Abstract
Increased ubiquitin-specific protease 5 (USP5) has been associated with tumorigenesis of malignancy including glioblastoma, melanoma and hepatocellular carcinoma. However, the role of USP5 in tumorigenesis of pancreatic ductal adenocarcinoma (PDAC) has not been studied yet. In this study, we demonstrated that USP5 was significantly upregulated in a panel of PDAC cell lines and correlated with FoxM1 protein expression. USP5 knockdown inhibited proliferation of PANC-1 and SW1990, two PDAC cell lines. In the mouse xenografted pancreatic tumor model, suppression of USP5 significantly decreased tumor growth, correlated with down regulation of FoxM1. Additionally, we found that overexpression of USP5 stabilized the FoxM1 protein in PDAC cells. Overexpression of USP5 extended the half-life of FoxM1. Knockdown of USP5 in PANC-1 cells decreased FoxM1 protein level while the proteasome inhibitor MG-132 treatment restored FoxM1 expression. We also found that endogenous USP5 was coimmunoprecipitated with an endogenous FoxM1 from PANC-1 cells while FoxM1 was also coimmunoprecipitated with USP5. Furthermore, we also confirmed that USP5 regulated proliferation of PDAC via FoxM1 by rescuing the inhibitory effect of USP5 knockdown with ectopic expression of FoxM1 in USP5-depleted cells. Taken together, our study demonstrates that USP5 plays a critical role in tumorigenesis and progression of pancreatic cancer by stabilizing FoxM1 protein, and provides a rationale for USP5 being a potential therapeutic approach against PDAC.
Collapse
|
13
|
Song X, Fiati Kenston SS, Zhao J, Yang D, Gu Y. Roles of FoxM1 in cell regulation and breast cancer targeting therapy. Med Oncol 2017; 34:41. [PMID: 28176242 DOI: 10.1007/s12032-017-0888-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/12/2017] [Indexed: 10/25/2022]
Abstract
Forkhead box M1 (FoxM1) is an oncogenic transcription factor involved in a wide variety of cellular processes, such as cell cycle progression, proliferation, differentiation, migration, metabolism and DNA damage response. It is overexpressed in many human cancers, especially in breast cancers. Posttranslational modifications are known to play an important role in regulating the expression and transcriptional activity of FoxM1. In this review, we characterize the posttranslational modifications of FoxM1, summarize modifications of FoxM1 by different kinases, explore the relationship between the different sites of modifications and comprehensively describe how posttranslational modifications to regulate the function of FoxM1 by changing protein stability, nucleus localization and transcriptional activity. Additionally, we systematically summarize the roles of FoxM1 in breast cancer occurrence, therapy and drug resistance. The purpose of this paper tries to give a better understanding of the regulatory mechanisms of FoxM1 in cell regulation and highlights potential of a new method for breast cancer therapy by targeting FoxM1.
Collapse
Affiliation(s)
- Xin Song
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Samuel Selorm Fiati Kenston
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Jinshun Zhao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Danting Yang
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, People's Republic of China.
| | - Yuanliang Gu
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, 818 Fenghua Road, Ningbo, 315211, Zhejiang, People's Republic of China.
| |
Collapse
|
14
|
Panaccione A, Zhang Y, Mi Y, Mitani Y, Yan G, Prasad ML, McDonald WH, El-Naggar AK, Yarbrough WG, Ivanov SV. Chromosomal abnormalities and molecular landscape of metastasizing mucinous salivary adenocarcinoma. Oral Oncol 2017; 66:38-45. [PMID: 28249646 DOI: 10.1016/j.oraloncology.2016.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/07/2016] [Accepted: 12/14/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Mucinous adenocarcinoma of the salivary gland (MAC) is a lethal cancer with unknown molecular etiology and a high propensity to lymph node metastasis. Mostly due to its orphan status, MAC remains one of the least explored cancers that lacks cell lines and mouse models that could help translational and pre-clinical studies. Surgery with or without radiation remains the only treatment modality but poor overall survival (10-year, 44%) underscores the urgent need for mechanism-based therapies. METHODS We developed the first patient-derived xenograft (PDX) model for pre-clinical MAC studies and a cell line that produces aggressively growing tumors after subcutaneous injection into nude mice. We performed cytogenetic, exome, and proteomic profiling of MAC to identify driving mutations, therapeutic targets, and pathways involved in aggressive cancers based on TCGA database mining and GEO analysis. RESULTS We identified in MAC KRAS (G13D) and TP53 (R213X) mutations that have been previously reported as drivers in a variety of highly aggressive cancers. Somatic mutations were also found in KDM6A, KMT2D, and other genes frequently mutated in colorectal and other cancers: FAT1, NBEA, RELN, RLP1B, and ZFHX3. Proteomic analysis of MAC implied epigenetic up-regulation of a genetic program involved in proliferation and cancer stem cell maintenance. CONCLUSION Genomic and proteomic analyses provided the first insight into potential molecular drivers of MAC metastases pointing at common mechanisms of CSC propagation in aggressive cancers. The in vitro/in vivo models that we created should aid in the development and validation of new treatment strategies against MAC.
Collapse
Affiliation(s)
- Alex Panaccione
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, 789 Howard Avenue, New Haven, CT 06519, USA
| | - Yi Zhang
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, 789 Howard Avenue, New Haven, CT 06519, USA
| | - Yanfang Mi
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, 789 Howard Avenue, New Haven, CT 06519, USA
| | - Yoshitsugu Mitani
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Guo Yan
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Manju L Prasad
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - W Hayes McDonald
- Proteomics Laboratory, Mass Spectrometry Research Center, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Adel K El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA; Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Wendell G Yarbrough
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, 789 Howard Avenue, New Haven, CT 06519, USA; H&N Disease Center, Smilow Cancer Hospital, New Haven, CT, USA; Molecular Virology Program, Yale Cancer Center, New Haven, CT, USA
| | - Sergey V Ivanov
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, 789 Howard Avenue, New Haven, CT 06519, USA.
| |
Collapse
|
15
|
Nestal de Moraes G, Bella L, Zona S, Burton MJ, Lam EWF. Insights into a Critical Role of the FOXO3a-FOXM1 Axis in DNA Damage Response and Genotoxic Drug Resistance. Curr Drug Targets 2016; 17:164-77. [PMID: 25418858 PMCID: PMC5403963 DOI: 10.2174/1389450115666141122211549] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 11/06/2014] [Accepted: 11/19/2014] [Indexed: 11/22/2022]
Abstract
FOXO3a and FOXM1 are two forkhead transcription factors with antagonistic roles in cancer and DNA damage response. FOXO3a functions like a typical tumour suppressor, whereas FOXM1 is a potent oncogene aberrantly overexpressed in genotoxic resistant cancers. FOXO3a not only represses FOXM1 expression but also its transcriptional output. Recent research has provided novel insights into a central role for FOXO3a and FOXM1 in DNA damage response. The FOXO3a-FOXM1 axis plays a pivotal role in DNA damage repair and the accompanied cellular response through regulating the expression of genes essential for DNA damage sensing, mediating, signalling and repair as well as for senescence, cell cycle and cell death control. In this manner, the FOXO3a-FOXM1 axis also holds the key to cell fate decision in response to genotoxic therapeutic agents and controls the equilibrium between DNA repair and cell termination by cell death or senescence. As a consequence, inhibition of FOXM1 or reactivation of FOXO3a in cancer cells could enhance the efficacy of DNA damaging cancer therapies by decreasing the rate of DNA repair and cell survival while increasing senescence and cell death. Conceptually, targeting FOXO3a and FOXM1 may represent a promising molecular therapeutic option for improving the efficacy and selectivity of DNA damage agents, particularly in genotoxic agent resistant cancer. In addition, FOXO3a, FOXM1 and their downstream transcriptional targets may also be reliable diagnostic biomarkers for predicting outcome, for selecting therapeutic options, and for monitoring treatments in DNA-damaging agent therapy.
Collapse
Affiliation(s)
| | | | | | | | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
16
|
Zona S, Bella L, Burton MJ, Nestal de Moraes G, Lam EWF. FOXM1: an emerging master regulator of DNA damage response and genotoxic agent resistance. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:1316-22. [PMID: 25287128 PMCID: PMC4316173 DOI: 10.1016/j.bbagrm.2014.09.016] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/07/2014] [Accepted: 09/25/2014] [Indexed: 02/03/2023]
Abstract
FOXM1 is a transcription factor required for a wide spectrum of essential biological functions, including DNA damage repair, cell proliferation, cell cycle progression, cell renewal, cell differentiation and tissue homeostasis. Recent evidence suggests that FOXM1 also has a role in many aspects of the DNA damage response. Accordingly, FOXM1 drives the transcription of genes for DNA damage sensors, mediators, signal transducers and effectors. As a result of these functions, it plays an integral part in maintaining the integrity of the genome and so is key to the propagation of accurate genetic information to the next generation. Preserving the genetic code is a vital means of suppressing cancer and other genetic diseases. Conversely, FOXM1 is also a potent oncogenic factor that is essential for cancer initiation, progression and drug resistance. An enhanced FOXM1 DNA damage repair gene expression network can confer resistance to genotoxic agents. Developing a thorough understanding of the regulation and function of FOXM1 in DNA damage response will improve the diagnosis and treatment of diseases including cancer, neurodegenerative conditions and immunodeficiency disorders. It will also benefit cancer patients with acquired genotoxic agent resistance. FOXM1 is a potent oncogenic factor essential for cancer initiation, progression and drug resistance. FOXM1 also drives the transcription of genes for DNA damage sensors, mediators, signal transducers and effectors. It plays an integral part in maintaining the integrity of the genome. An enhanced FOXM1 DNA damage repair gene expression network can confer resistance to genotoxic agents.
Collapse
Affiliation(s)
- Stefania Zona
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Laura Bella
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Matthew J Burton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Gabriela Nestal de Moraes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.
| |
Collapse
|
17
|
Singh PK, Srivastava AK, Rath SK, Dalela D, Goel MM, Bhatt MLB. Expression and clinical significance of Centrosomal protein 55 (CEP55) in human urinary bladder transitional cell carcinoma. Immunobiology 2014; 220:103-8. [PMID: 25178936 DOI: 10.1016/j.imbio.2014.08.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 08/14/2014] [Indexed: 02/07/2023]
Abstract
Bladder cancer (BC) is one among the most common and lethal urothelial malignancies worldwide. The expression of cancer-testis (CT) antigens in some tumours and restricted expression among normal tissues make CT antigens as attractive vaccine targets. In this context, we evaluated Centrosomal protein 55 kDa (CEP55), which is specifically expressed in normal human testis and various malignancies. Until the expression pattern of CEP55 in transitional cell carcinoma (TCC) of human urinary bladder and its clinical significance are not known. The aim of the present study is to evaluate mRNA/protein expression of CEP55 in TCCs of urinary bladder and correlate its expression with the clinicopathological characteristics of BC patients. In this study, the methods of quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) were used to investigate mRNA/protein expression of CEP55 in TCC. Independent Student's t test, ANOVA and Chi-square (χ(2)) were used to analyze the data statistically. We observed CEP55 mRNA overexpression in testis and 48.7% of BC patients. Relative mean fold expression of CEP55 mRNA was found to be significantly (p<0.01) higher in muscle-invasive bladder cancer (MIBC) as compared to non-muscle-invasive bladder cancer (NMIBC) patients (7.88±3.88 vs. 4.75±2.30, p=0.01). CEP55 protein expression was evaluated using IHC and cytoplasmic staining pattern was recorded in formalin fixed, paraffin-embedded (FFPE) bladder tumour tissues. No significant difference was observed in protein expression of CEP55 between the two groups (NMIBC and MIBC patients) (72.2% vs. 69.0%, p=0.774). No significant protein expression of CEP55 was observed among adjacent noncancerous tissues (ANCTs) and benign prostatic hyperplasia (BPH) used as control. Our study results suggest that CEP55 mRNA/protein expression was observed is specific to TCC of human urinary bladder and might be used as a diagnostic biomarker and vaccine target in development of BC specific immunotherapy.
Collapse
Affiliation(s)
- P K Singh
- Department of Radiotherapy, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Anupam K Srivastava
- Department of Radiotherapy, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - S K Rath
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226001, India
| | - D Dalela
- Department of Urology, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - M M Goel
- Department of Pathology, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - M L B Bhatt
- Department of Radiation Oncology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh 226010, India.
| |
Collapse
|
18
|
Wierstra I. The transcription factor FOXM1 (Forkhead box M1): proliferation-specific expression, transcription factor function, target genes, mouse models, and normal biological roles. Adv Cancer Res 2013; 118:97-398. [PMID: 23768511 DOI: 10.1016/b978-0-12-407173-5.00004-2] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor, which stimulates cell proliferation and exhibits a proliferation-specific expression pattern. Accordingly, both the expression and the transcriptional activity of FOXM1 are increased by proliferation signals, but decreased by antiproliferation signals, including the positive and negative regulation by protooncoproteins or tumor suppressors, respectively. FOXM1 stimulates cell cycle progression by promoting the entry into S-phase and M-phase. Moreover, FOXM1 is required for proper execution of mitosis. Accordingly, FOXM1 regulates the expression of genes, whose products control G1/S-transition, S-phase progression, G2/M-transition, and M-phase progression. Additionally, FOXM1 target genes encode proteins with functions in the execution of DNA replication and mitosis. FOXM1 is a transcriptional activator with a forkhead domain as DNA binding domain and with a very strong acidic transactivation domain. However, wild-type FOXM1 is (almost) inactive because the transactivation domain is repressed by three inhibitory domains. Inactive FOXM1 can be converted into a very potent transactivator by activating signals, which release the transactivation domain from its inhibition by the inhibitory domains. FOXM1 is essential for embryonic development and the foxm1 knockout is embryonically lethal. In adults, FOXM1 is important for tissue repair after injury. FOXM1 prevents premature senescence and interferes with contact inhibition. FOXM1 plays a role for maintenance of stem cell pluripotency and for self-renewal capacity of stem cells. The functions of FOXM1 in prevention of polyploidy and aneuploidy and in homologous recombination repair of DNA-double-strand breaks suggest an importance of FOXM1 for the maintenance of genomic stability and chromosomal integrity.
Collapse
|
19
|
Wierstra I. FOXM1 (Forkhead box M1) in tumorigenesis: overexpression in human cancer, implication in tumorigenesis, oncogenic functions, tumor-suppressive properties, and target of anticancer therapy. Adv Cancer Res 2013; 119:191-419. [PMID: 23870513 DOI: 10.1016/b978-0-12-407190-2.00016-2] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor and is also intimately involved in tumorigenesis. FOXM1 stimulates cell proliferation and cell cycle progression by promoting the entry into S-phase and M-phase. Additionally, FOXM1 is required for proper execution of mitosis. In accordance with its role in stimulation of cell proliferation, FOXM1 exhibits a proliferation-specific expression pattern and its expression is regulated by proliferation and anti-proliferation signals as well as by proto-oncoproteins and tumor suppressors. Since these factors are often mutated, overexpressed, or lost in human cancer, the normal control of the foxm1 expression by them provides the basis for deregulated FOXM1 expression in tumors. Accordingly, FOXM1 is overexpressed in many types of human cancer. FOXM1 is intimately involved in tumorigenesis, because it contributes to oncogenic transformation and participates in tumor initiation, growth, and progression, including positive effects on angiogenesis, migration, invasion, epithelial-mesenchymal transition, metastasis, recruitment of tumor-associated macrophages, tumor-associated lung inflammation, self-renewal capacity of cancer cells, prevention of premature cellular senescence, and chemotherapeutic drug resistance. However, in the context of urethane-induced lung tumorigenesis, FOXM1 has an unexpected tumor suppressor role in endothelial cells because it limits pulmonary inflammation and canonical Wnt signaling in epithelial lung cells, thereby restricting carcinogenesis. Accordingly, FOXM1 plays a role in homologous recombination repair of DNA double-strand breaks and maintenance of genomic stability, that is, prevention of polyploidy and aneuploidy. The implication of FOXM1 in tumorigenesis makes it an attractive target for anticancer therapy, and several antitumor drugs have been reported to decrease FOXM1 expression.
Collapse
|
20
|
Yan W, Shih J, Rodriguez-Canales J, Tangrea MA, Player A, Diao L, Hu N, Goldstein AM, Wang J, Taylor PR, Lippman SM, Wistuba II, Emmert-Buck MR, Erickson HS. Three-dimensional mRNA measurements reveal minimal regional heterogeneity in esophageal squamous cell carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 182:529-39. [PMID: 23219752 DOI: 10.1016/j.ajpath.2012.10.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 09/24/2012] [Accepted: 10/15/2012] [Indexed: 12/11/2022]
Abstract
The classic tumor clonal evolution theory postulates that cancers change over time to produce unique molecular subclones within a parent neoplasm, presumably including regional differences in gene expression. More recently, however, this notion has been challenged by studies showing that tumors maintain a relatively stable transcript profile. To examine these competing hypotheses, we microdissected discrete subregions containing approximately 3000 to 8000 cells (500 to 1500 μm in diameter) from ex vivo esophageal squamous cell carcinoma (ESCC) specimens and analyzed transcriptomes throughout three-dimensional tumor space. Overall mRNA profiles were highly similar in all 59 intratumor comparisons, in distinct contrast to the markedly different global expression patterns observed in other dissected cell populations. For example, normal esophageal basal cells contained 1918 and 624 differentially expressed genes at a greater than twofold level (95% confidence level of <5% false positives), compared with normal differentiated esophageal cells and ESCC, respectively. In contrast, intratumor regions had only zero to four gene changes at a greater than twofold level, with most tumor comparisons showing none. The present data indicate that, when analyzed using a standard array-based method at this level of histological resolution, ESCC contains little regional mRNA heterogeneity.
Collapse
Affiliation(s)
- Wusheng Yan
- Pathogenetics Unit, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Halasi M, Gartel AL. Targeting FOXM1 in cancer. Biochem Pharmacol 2012; 85:644-652. [PMID: 23103567 DOI: 10.1016/j.bcp.2012.10.013] [Citation(s) in RCA: 358] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/15/2012] [Accepted: 10/17/2012] [Indexed: 11/28/2022]
Abstract
Oncogenic transcription factor FOXM1 is overexpressed in the majority of human cancers. In addition, FOXM1 has been implicated in cell migration, invasion, angiogenesis and metastasis. The important role of FOXM1 in cancer affirms its significance for therapeutic intervention. Current data suggest that targeting FOXM1 in mono- or combination therapy may have promising therapeutic benefits for the treatment of cancer. However, challenges with the delivery of anti-FOXM1 siRNA to tumors and the absence of small molecules, which specifically inhibit FOXM1, are delaying the development of FOXM1 inhibitors as feasible anticancer drugs. In this review, we describe and summarize the efforts that have been made to target FOXM1 in cancer and the consequences of FOXM1 suppression in human cancer cells.
Collapse
Affiliation(s)
- Marianna Halasi
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Andrei L Gartel
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|