1
|
Miller AL, Garcia PL, Vance RB, Heard EO, Brown EJ, Yoon KJ. The BET inhibitor sensitivity is associated with the expression level of CDC25B in pancreatic cancer models. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:40. [PMID: 39534870 PMCID: PMC11555179 DOI: 10.20517/cdr.2024.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Aim: Cell division cycle 25B (CDC25B) belongs to the CDC25 family of phosphatases that regulate cell cycle progression. CDC25B also contributes to tumor initiation and progression, but no connection between CDC25B levels and drug sensitivity in pancreatic cancer has been reported. Based on our finding that bromodomain and extraterminal domain (BET) inhibitors decrease levels of CDC25B, we aim to compare the sensitivity of models expressing contrasting levels of CDC25B to the BET inhibitor JQ1, in pancreatic cancer cell lines in vitro and in patient-derived xenograft (PDX) models of pancreatic ductal adenocarcinoma (PDAC) in vivo. Methods: We compared the efficacy of the standard of care agent gemcitabine with the BET inhibitor JQ1, using alamarBlue assays to determine IC50s of three pancreatic cancer cell lines in vitro. We used immunohistochemistry (IHC) and immunoblot (IB) to detect CDC25B. We also compared the effect of each agent on the progression of PDX models of PDAC in vivo with contrasting levels of CDC25B. Results: Immunohistochemical data demonstrated that levels of CDC25B differed by ~2- to 5-fold in cell lines and PDX models used. In vitro data showed that the level of CDC25B paralleled sensitivity to JQ1. Similarly, in vivo data showed that tumors with high-level CDC25B were more sensitive to JQ1 than tumors with lower CDC25B. The combination of JQ1 + a pan CDC25 inhibitor was synergistic in gemcitabine-resistant Panc1.gemR cells that had relatively high levels of CDC25B expression compared to parent cells. Conclusion: The data suggest that CDC25B may be an independent indicator of sensitivity to BET inhibitors and that CDC25B may contribute to gemcitabine insensitivity in this tumor type.
Collapse
Affiliation(s)
| | | | | | | | | | - Karina J. Yoon
- Department of Pharmacology and Toxicology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
2
|
Feng D, Lv J, Li K, Cao Q, Han J, Yu H, Cheng Y, Zhuang J, Cai L, Yang H, Yang X, Lu Q. CircZNF609 promotes bladder cancer progression and inhibits cisplatin sensitivity via miR-1200/CDC25B pathway. Cell Biol Toxicol 2023; 39:1-18. [PMID: 35567596 DOI: 10.1007/s10565-022-09715-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 04/07/2022] [Indexed: 12/21/2022]
Abstract
Circular RNAs (circRNAs) have been extensively studied in tumor development and treatment. CircZNF609 (hsa_circ_0000615) has been shown to serve as an oncogene in all kinds of solid tumors and may act as the novel biomarker in tumor diagnosis and therapy in tumor early diagnosis and therapy. However, the underlying character and mechanism of circZNF609 in cisplatin chemosensitivity and bladder cancer (BCa) development were unknown. The expression level of cell division cycle 25B (CDC25B), microRNA 1200 (miR-1200), and circZNF609 in BCa cells and tissues depended on quantitative real-time PCR (qRT-PCR). CDC25B protein level was assayed with Western blot. Functional assays in vitro and in vivo had been conducted to inspect the important role of circZNF609 on BCa progression and cisplatin chemosensitivity in BCa. RNA sequencing and online databases were used to predict the interactions among circZNF609, miR-1200, and CDC25B. Mechanistic exploration was confirmed by RNA pull-down assay, RNA fluorescence in situ hybridization (FISH) and Dual luciferase reporter assay. CircZNF609 expression was increased significantly in BCa cell lines and tissues. For BCa patients, increased expression of circZNF609 was correlated with a worse survival. In vitro and in vivo, enforced expression of circZNF609 enhanced BCa cells proliferation, migration, and cisplatin chemoresistance. Mechanistically, circZNF609 alleviated the inhibition effect on target CDC25B expression by sponging miR-1200. CircZNF609 promoted tumor growth through novel circZNF609/miR-1200/CDC25B axis, implying that circZNF609 has significant potential to act as a new diagnostic biomarker and therapeutic target in BCa. Enhancing cisplatin sensitivity is an important direction for bladder cancer management. 1. This research reveals that circZNF609 improves bladder cancer progression and inhibits cisplatin sensitivity by inducing G1/S cell cycle arrest via a novel miR-1200/CDC25B cascades. 2. CircZNF609 was confirmed associated with worse survival of bladder cancer patients. 3. CircZNF609 act as a prognostic biomarker for bladder cancer treatment.
Collapse
Affiliation(s)
- Dexiang Feng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jiancheng Lv
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Kai Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qiang Cao
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jie Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Yu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yidong Cheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Juntao Zhuang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Lingkai Cai
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Xiao Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Qiang Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
3
|
Molecular and Biological Investigation of Isolated Marine Fungal Metabolites as Anticancer Agents: A Multi-Target Approach. Metabolites 2023; 13:metabo13020162. [PMID: 36837781 PMCID: PMC9964656 DOI: 10.3390/metabo13020162] [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: 12/10/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Cancer is the leading cause of death globally, with an increasing number of cases being annually reported. Nature-derived metabolites have been widely studied for their potential programmed necrosis, cytotoxicity, and anti-proliferation leading to enrichment for the modern medicine, particularly within the last couple of decades. At a more rapid pace, the concept of multi-target agents has evolved from being an innovative approach into a regular drug development procedure for hampering the multi-fashioned pathophysiology and high-resistance nature of cancer cells. With the advent of the Red Sea Penicillium chrysogenum strain S003-isolated indole-based alkaloids, we thoroughly investigated the molecular aspects for three major metabolites: meleagrin (MEL), roquefortine C (ROC), and isoroquefortine C (ISO) against three cancer-associated biological targets Cdc-25A, PTP-1B, and c-Met kinase. The study presented, for the first time, the detailed molecular insights and near-physiological affinity for these marine indole alkaloids against the assign targets through molecular docking-coupled all-atom dynamic simulation analysis. Findings highlighted the superiority of MEL's binding affinity/stability being quite in concordance with the in vitro anticancer activity profile conducted via sulforhodamine B bioassay on different cancerous cell lines reaching down to low micromolar or even nanomolar potencies. The advent of lengthy structural topologies via the metabolites' extended tetracyclic cores and aromatic imidazole arm permitted multi-pocket accommodation addressing the selectivity concerns. Additionally, the presence decorating polar functionalities on the core hydrophobic tetracyclic ring contributed compound's pharmacodynamic preferentiality. Introducing ionizable functionality with more lipophilic characters was highlighted to improve binding affinities which was also in concordance with the conducted drug-likeness/pharmacokinetic profiling for obtaining a balanced pharmacokinetic/dynamic profile. Our study adds to the knowledge regarding drug development and optimization of marine-isolated indole-based alkaloids for future iterative synthesis and pre-clinical investigations as multi-target anticancer agents.
Collapse
|
4
|
In Silico Identification of Small Molecules as New Cdc25 Inhibitors through the Correlation between Chemosensitivity and Protein Expression Pattern. Int J Mol Sci 2021; 22:ijms22073714. [PMID: 33918281 PMCID: PMC8038176 DOI: 10.3390/ijms22073714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/11/2023] Open
Abstract
The cell division cycle 25 (Cdc25) protein family plays a crucial role in controlling cell proliferation, making it an excellent target for cancer therapy. In this work, a set of small molecules were identified as Cdc25 modulators by applying a mixed ligand-structure-based approach and taking advantage of the correlation between the chemosensitivity of selected structures and the protein expression pattern of the proposed target. In the first step of the in silico protocol, a set of molecules acting as Cdc25 inhibitors were identified through a new ligand-based protocol and the evaluation of a large database of molecular structures. Subsequently, induced-fit docking (IFD) studies allowed us to further reduce the number of compounds biologically screened. In vitro antiproliferative and enzymatic inhibition assays on the selected compounds led to the identification of new structurally heterogeneous inhibitors of Cdc25 proteins. Among them, J3955, the most active inhibitor, showed concentration-dependent antiproliferative activity against HepG2 cells, with GI50 in the low micromolar range. When J3955 was tested in cell-cycle perturbation experiments, it caused mitotic failure by G2/M-phase cell-cycle arrest. Finally, Western blotting analysis showed an increment of phosphorylated Cdk1 levels in cells exposed to J3955, indicating its specific influence in cellular pathways involving Cdc25 proteins.
Collapse
|
5
|
Parajón E, Surcel A, Robinson DN. The mechanobiome: a goldmine for cancer therapeutics. Am J Physiol Cell Physiol 2020; 320:C306-C323. [PMID: 33175572 DOI: 10.1152/ajpcell.00409.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer progression is dependent on heightened mechanical adaptation, both for the cells' ability to change shape and to interact with varying mechanical environments. This type of adaptation is dependent on mechanoresponsive proteins that sense and respond to mechanical stress, as well as their regulators. Mechanoresponsive proteins are part of the mechanobiome, which is the larger network that constitutes the cell's mechanical systems that are also highly integrated with many other cellular systems, such as gene expression, metabolism, and signaling. Despite the altered expression patterns of key mechanobiome proteins across many different cancer types, pharmaceutical targeting of these proteins has been overlooked. Here, we review the biochemistry of key mechanoresponsive proteins, specifically nonmuscle myosin II, α-actinins, and filamins, as well as the partnering proteins 14-3-3 and CLP36. We also examined a wide range of data sets to assess how gene and protein expression levels of these proteins are altered across many different cancer types. Finally, we determined the potential of targeting these proteins to mitigate invasion or metastasis and suggest that the mechanobiome is a goldmine of opportunity for anticancer drug discovery and development.
Collapse
Affiliation(s)
- Eleana Parajón
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexandra Surcel
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
6
|
Ding Y, Cao Q, Wang C, Duan H, Shen H. LGALS4 as a Prognostic Factor in Urothelial Carcinoma of Bladder Affects Cell Functions. Technol Cancer Res Treat 2020; 18:1533033819876601. [PMID: 31558111 PMCID: PMC6767717 DOI: 10.1177/1533033819876601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND To identify the hub genes related to urothelial carcinoma of the bladder prognosis and to understand their underlying mechanism. METHODS The expression profiles of 18 pairs of urothelial carcinoma of the bladder patient tissue and paired adjacent tissue obtained from the Cancer Genome Atlas were performed. Weighted gene coexpression network analysis was employed to screen gene modules and hub genes with significant differential expressions in urothelial carcinoma of the bladder. The hub genes expression in urothelial carcinoma of the bladder tissues was validated by reverse transcription-quantitative polymerase chain reaction. The overall survival curve and disease-free survival curve of prognostic factor (LGALS4) were plotted using the Kaplan-Meier method. Furthermore, LGALS4 messenger RNA and protein expression were also assessed in 2 urothelial carcinoma of the bladder cell lines (T24 and 5637) by quantitative reverse transcription-polymerase chain reaction and Western blot. The functions of urothelial carcinoma of the bladder cells with transfected pcDNA3.1-LGALS4 were identified through MTT assay, plate clone formation assay, flow cytometry, and cell migration experiments. RESULTS LGALS4 was the hub gene of pink module and it was related to prognosis. Higher LGALS4 expression predicted higher probabilities of overall survival and disease-free survival. Overexpression of LGALS4 in urothelial carcinoma of the bladder cells suppressed cell viability and migration but induced apoptosis. CONCLUSION LGALS4 played a critical role in the progression of urothelial carcinoma of the bladder and held a promise to be the biomarker for diagnosis and treatment of urothelial carcinoma of the bladder. It predicted good prognosis of urothelial carcinoma of the bladder and restrained the growth and migration of urothelial carcinoma of the bladder cells.
Collapse
Affiliation(s)
- Yu Ding
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qifeng Cao
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chen Wang
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Huangqi Duan
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Haibo Shen
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
7
|
Giulietti M, Occhipinti G, Righetti A, Bracci M, Conti A, Ruzzo A, Cerigioni E, Cacciamani T, Principato G, Piva F. Emerging Biomarkers in Bladder Cancer Identified by Network Analysis of Transcriptomic Data. Front Oncol 2018; 8:450. [PMID: 30370253 PMCID: PMC6194189 DOI: 10.3389/fonc.2018.00450] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/25/2018] [Indexed: 01/03/2023] Open
Abstract
Bladder cancer is a very common malignancy. Although new treatment strategies have been developed, the identification of new therapeutic targets and reliable diagnostic/prognostic biomarkers for bladder cancer remains a priority. Generally, they are found among differentially expressed genes between patients and healthy subjects or among patients with different tumor stages. However, the classical approach includes processing these data taking into consideration only the expression of each single gene regardless of the expression of other genes. These complex gene interaction networks can be revealed by a recently developed systems biology approach called Weighted Gene Co-expression Network Analysis (WGCNA). It takes into account the expression of all genes assessed in an experiment in order to reveal the clusters of co-expressed genes (modules) that, very probably, are also co-regulated. If some genes are co-expressed in controls but not in pathological samples, it can be hypothesized that a regulatory mechanism was altered and that it could be the cause or the effect of the disease. Therefore, genes within these modules could play a role in cancer and thus be considered as potential therapeutic targets or diagnostic/prognostic biomarkers. Here, we have reviewed all the studies where WGCNA has been applied to gene expression data from bladder cancer patients. We have shown the importance of this new approach in identifying candidate biomarkers and therapeutic targets. They include both genes and miRNAs and some of them have already been identified in the literature to have a role in bladder cancer initiation, progression, metastasis, and patient survival.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Giulia Occhipinti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandra Righetti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Massimo Bracci
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Alessandro Conti
- Department of Urology, Bressanone/Brixen Hospital, Bressanone, Italy
| | - Annamaria Ruzzo
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Fano, Italy
| | - Elisabetta Cerigioni
- Unit of Pediatric and Specialistic Surgery, United Hospitals, "G.Salesi", Ancona, Italy
| | - Tiziana Cacciamani
- Department of Life and Environmental Science, Polytechnic University of Marche, Ancona, Italy
| | - Giovanni Principato
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
8
|
Liu S, Jiang H, Wen H, Ding Q, Feng C. Knockdown of tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ) enhances tumorigenesis both in vivo and in vitro in bladder cancer. Oncol Rep 2018; 39:2127-2135. [PMID: 29512774 PMCID: PMC5928769 DOI: 10.3892/or.2018.6294] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 02/28/2018] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer is the most common tumor of the urinary tract. Tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ), a gene encoding the 14-3-3ζ protein, has been observed to be frequently amplified in bladder cancer. However, the role of 14-3-3ζ in various types of cancer is controversial. With reproduction of The Cancer Genome Atlas database, we searched the correlation of YWHAZ expression with survival outcomes of multiple cancers in silico. Our results revealed that only in bladder cancer was there a positive survival trend with YWHAZ overexpression. To study its role in bladder cancer, YWHAZ was successfully downregulated by lentivirus in 5637 and T24 cells. MTT and colony-formation assays showed that YWHAZ downregulation increased cell proliferation. Wound healing and Transwell assays showed that YWHAZ downregulation enhanced cell migration and invasiveness. FACS analysis showed that YWHAZ induced cell cycle arrest, but not apoptosis. A xenograft tumor model revealed that YWHAZ knockdown enhanced tumor growth. Gene set enrichment analysis confirmed that the cell cycle pathway plays a vital role in the function of the YWHAZ gene. In conclusion, knockdown of YWHAZ promoted both in vitro and in vivo tumorigenesis in bladder cancer and may be a novel biomarker for bladder cancer deserving further study.
Collapse
Affiliation(s)
- Shenghua Liu
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Haowen Jiang
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Hui Wen
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Qiang Ding
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Chenchen Feng
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
9
|
Bhargava S, Patil V, Shah RA, Somasundaram K. IGF2 mRNA binding protein 3 (IMP3) mediated regulation of transcriptome and translatome in glioma cells. Cancer Biol Ther 2017; 19:42-52. [PMID: 28485999 DOI: 10.1080/15384047.2017.1323601] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
RNA binding proteins mediate global regulation at the level of transcriptome and translatome of a cell. We studied the global level expression changes regulated by IMP3 in transcriptome and translatome by performing microarray using total cellular RNA and heavy polysome derived RNA of IMP3 silenced glioma cells respectively. Differentially regulated transcripts at the transcriptome level (n = 2388) and at the level of translatome (n = 479) were identified. Further, these transcripts were classified as direct and indirect targets on the basis of presence of IMP3 binding site. Additional investigation revealed that direct targets at transcriptome level were found to be associated with processes related to cell cycle, whereas direct targets at the translatome level participated in apoptosis related pathways. Probable mechanism of indirect regulation at both the levels is also investigated. Collectively, our study reveals multi-level gene expression regulation imposed by IMP3 in glioma cells.
Collapse
Affiliation(s)
- Shruti Bhargava
- a Department of Microbiology and Cell Biology , Indian Institute of Science , Bangalore , India
| | - Vikas Patil
- a Department of Microbiology and Cell Biology , Indian Institute of Science , Bangalore , India
| | - Riyaz Ahmad Shah
- a Department of Microbiology and Cell Biology , Indian Institute of Science , Bangalore , India
| | - Kumaravel Somasundaram
- a Department of Microbiology and Cell Biology , Indian Institute of Science , Bangalore , India
| |
Collapse
|
10
|
Hu J, Ye F, Cui M, Lee P, Wei C, Hao Y, Wang X, Wang Y, Lu Z, Galsky M, McBride R, Wang L, Wang D, Cordon-Cardo C, Wang C, Zhang DY. Protein Profiling of Bladder Urothelial Cell Carcinoma. PLoS One 2016; 11:e0161922. [PMID: 27626805 PMCID: PMC5023150 DOI: 10.1371/journal.pone.0161922] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
Abstract
This study aimed to detect protein changes that can assist to understand the underlying biology of bladder cancer. The data showed forty five proteins were found to be differentially expressed comparing tumors vs non-tumor tissues, of which EGFR and cdc2p34 were correlated with muscle invasion and histological grade. Ten proteins (ß-catenin, HSP70, autotaxin, Notch4, PSTPIP1, DPYD, ODC, cyclinB1, calretinin and EPO) were able to classify muscle invasive BCa (MIBC) into 2 distinct groups, with group 2 associated with poorer survival. Finally, 3 proteins (P2X7, cdc25B and TFIIH p89) were independent factors for favorable overall survival.
Collapse
Affiliation(s)
- Jinghai Hu
- Department of Urology, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Fei Ye
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Miao Cui
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Peng Lee
- Departments of Pathology, New York University, School of Medicine, New York, NY, 10010, United States of America
| | - Chengguo Wei
- Division of Nephrology, Department of Medicine, Mount Sinai School of Medicine, New York, NY, 10029, United States of America
| | - Yuanyuan Hao
- Department of Urology, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Xiaoqing Wang
- Department of Urology, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Yanbo Wang
- Department of Urology, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Zhihua Lu
- Department of Urology, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Matthew Galsky
- Division of Hematology and Oncology, Department of Medicine, Mount Sinai School of Medicine, New York, NY, 10029, United States of America
| | - Russell McBride
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Li Wang
- Departments of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY, 10029, United States of America
| | - Dongwen Wang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030002, China
| | - Carlos Cordon-Cardo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Chunxi Wang
- Department of Urology, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
- * E-mail: (DYZ); (CXW)
| | - David Y. Zhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
- * E-mail: (DYZ); (CXW)
| |
Collapse
|
11
|
Zhang Z, Zhang G, Kong C, Gong D. PP242 suppresses bladder cancer cell proliferation and migration through deactivating the mammalian target of rapamycin complex 2/AKT1 signaling pathway. Mol Med Rep 2015; 13:333-8. [PMID: 26548560 DOI: 10.3892/mmr.2015.4528] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 10/06/2015] [Indexed: 11/06/2022] Open
Abstract
While most cancer types are resistant to mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin, recent studies have identified mTORC2 as an important prospective therapeutic target for cancer. The present study assessed the effects of mTORC2 inhibitor PP242 on the proliferation and migration of bladder cancer cells by using Cell Counting Kit‑8, 5‑ethynyl‑2'‑deoxyuridine incorporation, wound healing and Transwell assays. Furthermore, the phosphorylation status of downstream signaling proteins of mTORC1 and mTORC2 was assessed using western blot analysis. The results demonstrated that PP242 concentration‑dependently inhibited the proliferation of bladder cancer cells. Simultaneously, the migration ability of bladder cancer cells was suppressed by PP242. In addition, PP242 markedly restrained the phosphorylation of AKT1 and mTORC2, while the phosphorylation status of S6K1 and mTORC1 was not affected. These results suggested that PP242 exerts potent inhibitory effects on bladder cancer cells by modulating the activity of the mTORC2/AKT1 pathway.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Guojun Zhang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, P.R. China
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Daxin Gong
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
12
|
Wang M, Zhu XY, Wang L, Lin Y. Expression and significance of CDC25B, PED/PEA-15 in esophageal carcinoma. Cancer Biother Radiopharm 2015; 30:139-45. [PMID: 25775393 DOI: 10.1089/cbr.2014.1701] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To explore the role of CDC25B, PED/PEA-15 in the development of esophageal carcinoma and its influence on the prognosis. METHODS Fluorescence quantitative real-time PCR and immunohistochemistry methods were used to analyze the expression of CDC25B, PED/PEA-15 in esophageal carcinoma. Moreover, survival analysis was done using the Kaplan-Meier method. RESULTS In 66 cases of esophageal cancer tissues, the relative content of CDC25B mRNA was 16.22 (13.93-18.90). The positive expression rate of CDC25B protein was 48.5%, significantly higher than normal mucosa tissues (0%) (p<0.01). The relative content of PED/PEA-15 mRNA was 12.47 (10.41-14.93). The positive expression rate of PED/PEA-15 protein was 68.2%, significantly higher than normal mucosa tissues (17.6%) (p<0.01). The CDC25B protein expression was correlated with differentiation grade and depth of invasion (p<0.05). The PED/PEA-15 protein expression was related to differentiation grade, lymph node metastasis, and depth of invasion (p<0.05). Survival analysis showed that the mean survival time of PED/PEA-15-positive expression group was lower compared with the negative expression group (χ(2)=5.549, p=0.018). Analysis of the relationship between CDC25B and PED/PEA-15 suggested that there was a positive correlation between them (r=4.061, p=0.044). CONCLUSION Both CDC25B and PED/PEA-15 play a certain role in the carcinogenesis of esophageal cancer, and PED/PEA-15 has a greater influence on postoperative survival time. They will be the new diagnostic/therapeutic targets in esophageal carcinoma.
Collapse
Affiliation(s)
- Ming Wang
- 1 Department of Radiation Therapy, Cangzhou Central Hospital , Cangzhou, China
| | | | | | | |
Collapse
|
13
|
Lund G, Dudkin S, Borkin D, Ni W, Grembecka J, Cierpicki T. Inhibition of CDC25B phosphatase through disruption of protein-protein interaction. ACS Chem Biol 2015; 10:390-4. [PMID: 25423142 PMCID: PMC4340349 DOI: 10.1021/cb500883h] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
CDC25 phosphatases are key cell cycle regulators and represent very attractive but challenging targets for anticancer drug discovery. Here, we explored whether fragment-based screening represents a valid approach to identify inhibitors of CDC25B. This resulted in identification of 2-fluoro-4-hydroxybenzonitrile, which directly binds to the catalytic domain of CDC25B. Interestingly, NMR data and the crystal structure demonstrate that this compound binds to the pocket distant from the active site and adjacent to the protein-protein interaction interface with CDK2/Cyclin A substrate. Furthermore, we developed a more potent analogue that disrupts CDC25B interaction with CDK2/Cyclin A and inhibits dephosphorylation of CDK2. Based on these studies, we provide a proof of concept that targeting CDC25 phosphatases by inhibiting their protein-protein interactions with CDK2/Cyclin A substrate represents a novel, viable opportunity to target this important class of enzymes.
Collapse
Affiliation(s)
- George Lund
- Department of Pathology, University of Michigan, 4510C MSRBI
1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5620, United States
| | - Sergii Dudkin
- Department of Pathology, University of Michigan, 4510C MSRBI
1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5620, United States
| | - Dmitry Borkin
- Department of Pathology, University of Michigan, 4510C MSRBI
1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5620, United States
| | - Wendi Ni
- Department of Pathology, University of Michigan, 4510C MSRBI
1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5620, United States
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, 4510C MSRBI
1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5620, United States
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, 4510C MSRBI
1150 West Medical Center Drive, Ann Arbor, Michigan 48109-5620, United States
| |
Collapse
|
14
|
Song GQ, Zhao Y. MicroRNA-211, a direct negative regulator of CDC25B expression, inhibits triple-negative breast cancer cells' growth and migration. Tumour Biol 2015; 36:5001-9. [PMID: 25680404 DOI: 10.1007/s13277-015-3151-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/26/2015] [Indexed: 10/24/2022] Open
Abstract
The non-coding microRNAs (miRNAs) have tissue- and disease-specific expression patterns. Dysregulation of miRNAs has been associated with initiation and progression of oncogenesis in humans. The abnormal expression of CDC25B phosphatases detected in a number of tumors implies that their dysregulation is involved in malignant transformation. Using miRNA target prediction software, we found that miR-211 could target the 3'UTR sequence of CDC25B. To shed light on their roles of miR-211 in breast cancer, the expression of miR-211 was examined by real-time RT-PCR in breast cancer and normal tissues. MiR-211 is significantly downregulated in breast cancer. MiR-211 re-expression suppressed cell growth, cell cycle, migration, and invasion in triple-negative breast cancer (TNBC) cell line MDA-MB231. Luciferase expression from a reporter vector containing the CDC25B -3'UTR was decreased when this construct was transfected with miR-211. The over-expression of miR-211 suppressed the endogenous CDC25B protein level in TNBC cells. For the first time, we demonstrate that miRNA-211 is a direct negative regulator of CDC25B expression in TNBC cells, alters other related target proteins CCNB1 and FOXM1, and then inhibits breast cancer cells growth, migration, and invasion and lead G2/M arrest. The transcriptional loss of miR-211 and the resultant increase in CDC25B expression facilitate increased genomic instability at an early stage of tumor development.
Collapse
Affiliation(s)
- Guo-qing Song
- Department of Pancreatic and Breast Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning, 110004, People's Republic of China
| | | |
Collapse
|
15
|
Li YL, Liu L, Xiao Y, Zeng T, Zeng C. 14-3-3σ is an independent prognostic biomarker for gastric cancer and is associated with apoptosis and proliferation in gastric cancer. Oncol Lett 2014; 9:290-294. [PMID: 25435977 PMCID: PMC4246703 DOI: 10.3892/ol.2014.2676] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 09/26/2014] [Indexed: 11/13/2022] Open
Abstract
14-3-3 proteins participate in various cellular processes, including apoptosis, proliferation and malignant transformation. 14-3-3σ, a member of the 14-3-3 protein family, is important in several types of cancer; however, little is known about the clinical significance and biological roles of 14-3-3σ in gastric cancer. The present study analyzed the expression pattern of 14-3-3σ in gastric cancer and investigated its correlation with the prognosis of gastric cancer patients. Furthermore, the association of 14-3-3σ with Ki-67, Bcl-2 and Bax was evaluated. 14-3-3σ was expressed at higher level in gastric cancer tissue compared with healthy gastric tissue, and 14-3-3σ expression was significantly correlated with tumor size and tumor node metastasis stage (P<0.05). To the best of our knowledge, the present study data are the first to suggest that 14-3-3σ expression has been significantly associated with poor prognosis in gastric cancer. Additionally, 14-3-3σ overexpression was positively correlated with Ki-67 and Bcl-2 expression levels. Thus, 14-3-3σ is a potential prognostic marker for gastric cancer patients, and may be involved in regulating the apoptosis and proliferation of gastric cancer cells.
Collapse
Affiliation(s)
- Yi-Liang Li
- Department of Neurology, The Central Hospital of Loudi Affiliated to the University of South China, Loudi, Hunan 417000, P.R. China
| | - Lihua Liu
- Department of Respiratory Disease, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yang Xiao
- Department of Orthopaedics, The Central Hospital of Loudi Affiliated to the University of South China, Loudi, Hunan 417000, P.R. China
| | - Tao Zeng
- School of Laboratory Medicine, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| | - Chao Zeng
- Department of Pathology, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
16
|
Vasamsetty L, Sahu D, Ganguly B, Khan FA, Mehta G. Total synthesis of novel bioactive natural product paracaseolide A and analogues: computational evaluation of a ‘proposed’ biomimetic Diels–Alder reaction. Tetrahedron 2014. [DOI: 10.1016/j.tet.2014.09.072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|