1
|
Shaldam MA, Almahli H, Angeli A, Badi RM, Khaleel EF, Zain-Alabdeen AI, Elsayed ZM, Elkaeed EB, Salem R, Supuran CT, Eldehna WM, Tawfik HO. Discovery of sulfonamide-tethered isatin derivatives as novel anticancer agents and VEGFR-2 inhibitors. J Enzyme Inhib Med Chem 2023; 38:2203389. [PMID: 37122176 PMCID: PMC10134960 DOI: 10.1080/14756366.2023.2203389] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
In this work, new isatin-based sulphonamides (6a-i, 11a-c, 12a-c) were designed and synthesised as potential dual VEGFR-2 and carbonic anhydrase inhibitors with anticancer activities. Firstly, all target isatins were examined for in vitro antitumor action on NCI-USA panel (58 tumour cell lines). Then, the most potent derivatives were examined for the potential CA inhibitory action towards the physiologically relevant hCA isoforms I, II, and tumour-linked hCA IX isoform, in addition, the VEGFR-2 inhibitory activity was evaluated. The target sulphonamides failed to inhibit the CA isoforms that could be attributable to the steric effect of the neighbouring methoxy group, whereas they displayed potent VEGFR-2 inhibitory effect. Following that, isatins 11b and 12b were tested for their influence on the cell cycle disturbance, and towards the apoptotic potential. Finally, detailed molecular modelling analyses, including docking and molecular dynamics, were carried out to assess the binding mode and stability of target isatins.
Collapse
Affiliation(s)
- Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Hadia Almahli
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Andrea Angeli
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Rehab Mustafa Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | | | - Zainab M Elsayed
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh Uinversity, Kafrelsheikh, Egypt
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Rofaida Salem
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
- School of Biotechnology, Badr University in Cairo, Badr City, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
2
|
Decidua Parietalis Mesenchymal Stem/Stromal Cells and Their Secretome Diminish the Oncogenic Properties of MDA231 Cells In Vitro. Cells 2021; 10:cells10123493. [PMID: 34944000 PMCID: PMC8700435 DOI: 10.3390/cells10123493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been shown to suppress tumor growth, inhibit angiogenesis, regulate cellular signaling, and induce apoptosis in cancer cells. We have earlier reported that placenta-derived decidua parietalis mesenchymal stem/stromal cells (DPMSCs) not only retained their functional characteristics in the cancer microenvironment but also exhibited increased expression of anti-apoptotic genes, demonstrating their anti-tumor properties in the tumor setting. In this study, we have further evaluated the effects of DPMSCs on the functional outcome of human breast cancer cell line MDA231. MDA231 cells were exposed to DPMSCs, and their biological functions, including adhesion, proliferation, migration, and invasion, were evaluated. In addition, genomic and proteomic modifications of the MDA231 cell line, in response to the DPMSCs, were also evaluated. MDA231 cells exhibited a significant reduction in proliferation, migration, and invasion potential after their treatment with DPMSCs. Furthermore, DPMSC treatment diminished the angiogenic potential of MDA231 cells. DPMSC treatment modulated the expression of various pro-apoptotic as well as oncogenes in MDA231 cells. The properties of DPMSCs to inhibit the invasive characteristics of MDA231 cells demonstrate that they may be a useful candidate in a stem-cell-based therapy against cancer.
Collapse
|
3
|
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance. Cancers (Basel) 2020; 12:E3323. [PMID: 33182737 PMCID: PMC7697134 DOI: 10.3390/cancers12113323] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The taxane family of chemotherapy drugs has been used to treat a variety of mostly epithelial-derived tumors and remain the first-line treatment for some cancers. Despite the improved survival time and reduction of tumor size observed in some patients, many have no response to the drugs or develop resistance over time. Taxane resistance is multi-faceted and involves multiple pathways in proliferation, apoptosis, metabolism, and the transport of foreign substances. In this review, we dive deeper into hypothesized resistance mechanisms from research during the last decade, with a focus on the cancer types that use taxanes as first-line treatment but frequently develop resistance to them. Furthermore, we will discuss current clinical inhibitors and those yet to be approved that target key pathways or proteins and aim to reverse resistance in combination with taxanes or individually. Lastly, we will highlight taxane response biomarkers, specific genes with monitored expression and correlated with response to taxanes, mentioning those currently being used and those that should be adopted. The future directions of taxanes involve more personalized approaches to treatment by tailoring drug-inhibitor combinations or alternatives depending on levels of resistance biomarkers. We hope that this review will identify gaps in knowledge surrounding taxane resistance that future research or clinical trials can overcome.
Collapse
Affiliation(s)
- Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Camden A. Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Lorena V. Morejon-Lasso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| |
Collapse
|
4
|
Karna SKL, Lone BA, Ahmad F, Shahi N, Pokharel YR. Knockdown of CSNK2ß suppresses MDA-MB231 cell growth, induces apoptosis, inhibits migration and invasion. EXCLI JOURNAL 2020; 19:1211-1226. [PMID: 33013272 PMCID: PMC7527516 DOI: 10.17179/excli2020-2363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common cancer among women worldwide. Among different types of breast cancer known, treatment of triple-negative breast cancer is a major challenge because of its aggressiveness and poor prognosis; thus, identification of specific drivers is required for targeted therapies of breast cancer malignancy. Protein Casein Kinase (CSNK) is a serine/threonine kinase that exists as a tetrameric complex consisting of two catalytic (α and /or α') and two regulatory β subunits. CSNK2β can also function independently without catalytic subunits and exist as a distinct population in cells. This study aims to elucidate the role of Casein Kinase 2β (CSNK2β) gene in cell proliferation, cell cycle, migration and apoptosis of triple-negative breast cancer MDA-MB-231 cells. The silencing of CSNK2β in MDA-MB-231 cells resulted in decreased cell viability and colony formation. Cell cycle analysis showed a significant arrest of cells in G2M phase. Hoechst and CM-H2DCFDA staining showed nuclear condensation and augmented intracellular reactive oxygen species (ROS) production. Furthermore, silencing of CSNK2β in MDA-MB-231 cells modulated the apoptotic machinery- BAX, Bcl-xL, and caspase 3; autophagy machinery-Beclin-1 and LC3-1; and inhibited the vital markers (p-ERK, c-Myc, NF-κB, E2F1, PCNA, p38-α) associated with cell proliferation and DNA replication pathways. In addition, knockdown of CSNK2β also affected the migration potential of MDA-MB-231, as observed in the wound healing and transwell migration assays. Altogether, the study suggests that CSNK2β silencing may offer future therapeutic target in triple-negative breast cancer.
Collapse
Affiliation(s)
- Shibendra Kumar Lal Karna
- Faculty of Life Science and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi-110021, India
| | - Bilal Ahmad Lone
- Faculty of Life Science and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi-110021, India
| | - Faiz Ahmad
- Faculty of Life Science and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi-110021, India
| | - Nerina Shahi
- Faculty of Life Science and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi-110021, India
| | - Yuba Raj Pokharel
- Faculty of Life Science and Biotechnology, South Asian University, Akbar Bhawan, Chanakyapuri, New Delhi-110021, India
| |
Collapse
|
5
|
Pirouzpanah S, Varshosaz P, Fakhrjou A, Montazeri V. The contribution of dietary and plasma folate and cobalamin to levels of angiopoietin-1, angiopoietin-2 and Tie-2 receptors depend on vascular endothelial growth factor status of primary breast cancer patients. Sci Rep 2019; 9:14851. [PMID: 31619709 PMCID: PMC6795805 DOI: 10.1038/s41598-019-51050-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 09/20/2019] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to determine the association of dietary folate and cobalamin with plasma levels of Angiopoietins (ANG), vascular endothelial growth factor-C (VEGF-C) and tyrosine kinase receptor-2 (Tie-2) of primary breast cancer patients. Women (n = 177), aged 30 to 75 years diagnosed with breast cancer were recruited from an ongoing case series study. Dietary intake of nutrients was estimated by using a validated food frequency questionnaire. Enzyme-linked immunosorbent assay was applied to measure biomarkers. MCF-7 cell cultures were supplemented with folic acid (0–40 μM) for 24 h to measure cell viability and fold change of expression by the real-time reverse transcriptase-polymerase chain reaction. Structural equation modeling was applied to analyze the structural relationships between the measured variables of nutrients and Angiopoietins. Dietary intake of folate and cobalamin showed a significant inverse correlation with plasma ANG-1 and ANG-2 (P < 0.05), particularly in subjects with estrogen-receptor positive tumors or low plasma VEGF-C. Plasma folate was positively associated with the ratio of ANG-1/ANG-2 (P < 0.05). Residual intake levels of total cobalamin were inversely associated with plasma ANG-1 when plasma stratum of VEGF-C was high (P < 0.05). Structural equation modeling identified a significant inverse contribution of folate profiles on the latent variable of Angiopoietins (coefficient β = −0.99, P < 0.05). Folic acid treatment resulted in dose-dependent down-regulations on ANGPT1 and ANGPT1/ANGPT2 ratio but VEGF and ANGPT2/VEGF were upregulated at folic acid >20 μM. Studying the contributing role of dietary folate to pro-angiogenic biomarkers in breast cancer patients can infer the preventive role of folate in the ANGs/VEGF-C-dependent cascade of tumor metastasis. By contrast, high concentrations of folic acid in vitro supported VEGF-C-dependent ANGPT2 overexpression might potentiate micro-lymphatic vessel development to support malignant cell dissemination.
Collapse
Affiliation(s)
- Saeed Pirouzpanah
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, 5166614711, Iran.
| | - Parisa Varshosaz
- Drug Applied Research Center/ and also Department of Biochemistry and Dietetics, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, 5166614711, Iran.,Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
| | - Ashraf Fakhrjou
- Department of Pathology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, 5156913193, Iran
| | - Vahid Montazeri
- Department of Thoracic Surgery, Faculty of Medicine, Surgery Ward, Tabriz University of Medical Sciences, and also Nour-Nejat Hospital, Tabriz, 5138665793, Iran
| |
Collapse
|
6
|
He X, Li S, Shi W, Lin Q, Ma J, Liu Y, Feng T, Cao X. Cyclin A1 is associated with poor prognosis in oesophageal squamous cell carcinoma. Oncol Lett 2019; 18:706-712. [PMID: 31289545 PMCID: PMC6546994 DOI: 10.3892/ol.2019.10377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of cyclin A1 (CCNA1) is implicated in the carcinogenesis, progression and metastasis of many types of solid tumours. In the present study, an mRNA single-channel expression profile chip experiment revealed that the CCNA1 mRNA levels in oesophageal squamous cell carcinoma (ESCC) were increased >10-fold compared with those in the adjacent non-cancer tissues. Reverse transcription-quantitative polymerase chain reaction and immunohistochemistry analyses were performed to additionally investigate the role of CCNA1 in the development and progression of ESCC in patients treated by radical resection of the oesophagus. The association between CCNA1 mRNA expression and the clinicopathological parameters of patients with ESCC was statistically analysed. The results indicated that upregulation of CCNA1 occurred in ~70% of patients with ESCC, and increased CCNA1 mRNA expression was significantly associated with advanced clinical stage, lymph node metastasis, invasiveness and poor clinical outcome, including disease-free survival and overall survival rates. Taken together, the data suggested that CCNA1 had an important function in ESCC development and progression, and may serve as a prognostic biomarker and therapeutic target in ESCC.
Collapse
Affiliation(s)
- Xiaoting He
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China.,Department of Medical Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214043, P.R. China
| | - Suqing Li
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Weihong Shi
- Department of Clinical Medicine, Yancheng Vocational Institute of Health Sciences, Yancheng, Jiangsu 224005, P.R. China
| | - Qingfeng Lin
- Department of Medical Oncology, Jiangyin People's Hospital, Jiangyin, Jiangsu 214400, P.R. China
| | - Jian Ma
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yang Liu
- Department of Pharmacy, No. 401 Hospital of Chinese People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Tingting Feng
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Xiufeng Cao
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China.,Department of Surgery, Nanjing Medical University, Taikang Xianlin Drum Tower Hospital, Jiangsu 210000, P.R. China
| |
Collapse
|
7
|
Cao J, Dong J, Wang Y, Chen Y. The expressions of DNA methyltransferase 1 (DNMT1) and cyclin A1 (CCNA1) in cervical carcinogenesis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:40-49. [PMID: 31933719 PMCID: PMC6943998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 11/26/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVES The purpose of the present study was to investigate the expressions of DNA methyltransferase 1 (DNMT1) and cyclin A1 (CCNA1) protein and mRNA in the process of cervical carcinogenesis. METHODS The specimens were separated into the following groups: control (n=30), low-grade squamous intraepithelial lesions (LSIL, n=30), high-grade squamous intraepithelial lesions (HSIL) and squamous cell cervical cancers (SCC, n=30). Immunohistochemical examination, Western blot and real-time quantitative PCR were used to investigate the protein and mRNA expressions of DNMT1 and CCNA1 in cervical tissues. RESULTS We found that the positive expression rate and intensity of DNMT1 mRNA and protein gradually increased in the process of cervical carcinogenesis. However, the increase of the positive expression rate of DNMT1 mRNA and protein form the LSIL group to HSIL group was not significant (P>0.05). After cervical intraepithelial neoplasia (CIN), the positive expression rate and intensity of CCNA1 mRNA and protein significantly decreased with the aggravation of cervical lesions (P<0.05), and there was no significant difference between the LSIL and control groups (P>0.05). CONCLUSIONS With the severity of cervical lesions, the expression of DNMT1 protein and mRNA increased gradually. The expression of CCNA1 protein and mRNA decreased gradually. The DNMT1 and CCNA1 expressions are associated with cervical lesions.
Collapse
Affiliation(s)
- Jiayu Cao
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital Tangshan, Hebei Province, China
| | - Jianxin Dong
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital Tangshan, Hebei Province, China
| | - Yan Wang
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital Tangshan, Hebei Province, China
| | - Yan Chen
- Department of Obstetrics and Gynecology, North China University of Science and Technology Affiliated Hospital Tangshan, Hebei Province, China
| |
Collapse
|
8
|
Sarwar M, Syed Khaja AS, Aleskandarany M, Karlsson R, Althobiti M, Ødum N, Mongan NP, Dizeyi N, Johnson H, Green AR, Ellis IO, Rakha EA, Persson JL. The role of PIP5K1α/pAKT and targeted inhibition of growth of subtypes of breast cancer using PIP5K1α inhibitor. Oncogene 2018; 38:375-389. [PMID: 30104711 PMCID: PMC6336681 DOI: 10.1038/s41388-018-0438-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/22/2018] [Accepted: 07/14/2018] [Indexed: 02/08/2023]
Abstract
Despite recent improvement in adjuvant therapies, triple-negative, and ER+ subtypes of breast cancer (BC) with metastatic potentials remain the leading cause of BC-related deaths. We investigated the role of phosphatidylinositol-4-phosphate 5-kinase alpha (PIP5Kα), a key upstream factor of PI3K/AKT, and the therapeutic effect of PIP5Kα inhibitor on subtypes of BC. The clinical importance of PIP5K1α and its association with survivals were analyzed using three BC cohorts from Nottingham (n = 913), KM plotter (n = 112) and TCGA (n = 817). Targeted overexpression or knockdown of PIP5K1α were introduced into BC cell lines. The effects of PIP5K1α and its inhibitor on growth and invasion of BC were confirmed by using in vitro assays including proliferation, migration, apoptosis and luciferase reporter assays and in vivo xenograft mouse models. All statistical tests were two-sided. PIP5K1α was associated with poor patient outcome in triple-negative BC (for PIP5K1α protein, p = 0.011 and for mRNA expression, p = 0.028, log-rank test). 29% of triple-negative BC had PIP5K1A gene amplification. Elevated level of PIP5K1α increased expression of pSer-473 AKT (p < 0.001) and invasiveness of triple-negative MDA-MB-231 cells (p < 0.001). Conversely, inhibition of PIP5K1α using its inhibitor ISA-2011B, or via knockdown suppressed growth and invasiveness of MDA-MB-231 xenografts (mean vehicle-treated controls = 2160 mm3, and mean ISA-2011B-treated = 600 mm3, p < 0.001). ISA-2011B-treatment reduced expression of pSer-473 AKT (p < 0.001) and its downstream effectors including cyclin D1, VEGF and its receptors, VEGFR1 and VEGFR2 (p < 0.001) in xenograft tumors. In ER+ cancer cells, PIP5K1α acted on pSer-473 AKT, and was in complexes with VEGFR2, serving as co-factor of ER-alpha to regulate activities of target genes including cyclin D1 and CDK1. Our study suggests that our developed PIP5K1α inhibitor has a great potential on refining targeted therapeutics for treatment of triple-negative and ER+ BC with abnormal PI3K/AKT pathways.
Collapse
Affiliation(s)
- Martuza Sarwar
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | | | - Mohammed Aleskandarany
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Richard Karlsson
- Division of Basal Tumor Biology, Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Maryam Althobiti
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Niels Ødum
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Nigel P Mongan
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom.,Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Nisthman Dizeyi
- Division of reproductive research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden
| | - Heather Johnson
- Department of Bio-Diagnosis, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Andrew R Green
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ian O Ellis
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Academic Pathology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jenny L Persson
- Division of Experimental Cancer Research, Department of Translational Medicine, Lund University, Clinical Research Centre, Malmö, Sweden. .,Division of Basal Tumor Biology, Department of Molecular Biology, Umeå University, Umeå, Sweden.
| |
Collapse
|
9
|
Jiang Z, Jiang J, Zhao B, Yang H, Wang Y, Guo S, Deng Y, Lu D, Ma T, Wang H, Wang J. CPNE1 silencing inhibits the proliferation, invasion and migration of human osteosarcoma cells. Oncol Rep 2017; 39:643-650. [PMID: 29207139 DOI: 10.3892/or.2017.6128] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/13/2017] [Indexed: 01/26/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignancy of the bone affecting children and adolescents. Copine 1 (CPNE1) is a highly conserved calcium-dependent phospholipid-binding protein and may function in regulating signal transduction and membrane trafficking. In the present study, we investigated CPNE1 expression in osteosarcoma tissues and cells, and studied the effects of small interfering RNA (siRNA)-targeting CPNE1 on proliferation, metastasis and chemosensitivity of the osteosarcoma cells. The results demonstrated that CPNE1 was highly expressed in the osteosarcoma tissues and cell lines. Moreover, functional investigations confirmed that CPNE1 knockdown significantly inhibited cell proliferation, colony formation, invasion and metastasis in Saos-2 and HOS cells. Western blot analysis indicated that CPNE1 silencing downregulated the expression of many proteins associated with tumorigenesis and development, including Ras, MEK-1/2, WNT1, β-catenin, cyclin A1, IRAK2 and cIAP2. In addition, CPNE1 downregulation enhanced the sensitivity of Saos-2 cells towards cisplatin and adriamycin. The present study provides deep insight into the clinical use of lentiviral-mediated CPNE1 silencing for osteosarcoma therapy.
Collapse
Affiliation(s)
- Zhenhuan Jiang
- Department of Orthopaedics, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, P.R. China
| | - Jiannong Jiang
- Department of Orthopaedics, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, P.R. China
| | - Bizeng Zhao
- Department of Orthopedics, Affiliated Sixth People's Hospital of Shanghai JiaoTong University, Shanghai 20023, P.R. China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| | - Yunliang Wang
- Department of Neurology, The 148 Central Hospital of PLA, Zibo, Shangdong 255000, P.R. China
| | - Shang Guo
- Department of Orthopedics, Affiliated Sixth People's Hospital of Shanghai JiaoTong University, Shanghai 20023, P.R. China
| | - Youping Deng
- Bioinformatics Core, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA
| | - Deyi Lu
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Tieliang Ma
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, P.R. China
| | - Hongwei Wang
- Shanghai Realgen Biotech Inc., Pudong New District, Shanghai 203215, P.R. China
| | - Jinzhi Wang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
10
|
Siddiqui ZI, Farooqui SR, Azam SA, Afroz M, Wajid S, Parveen S, Kazim SN. A comparative study of hepatitis B virus X protein mutants K130M, V131I and KV130/131MI to investigate their roles in fibrosis, cirrhosis and hepatocellular carcinoma. J Viral Hepat 2017; 24:1121-1131. [PMID: 28654219 DOI: 10.1111/jvh.12747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/06/2017] [Indexed: 01/04/2023]
Abstract
Hepatitis B virus (HBV) genomic mutations A1762T, G1764A and AG1762/1764TA cause production of HBV X protein (HBx) mutants, namely K130M, V131I and KV130/131MI. These mutations are important biomarkers for the development of cirrhosis and hepatocellular carcinoma (HCC) in chronic HBV patients. This study comparatively analyses the impact of intracellular expression of HBx mutants on HCC cell line Huh7. It was found that expression of KV130/131MI induced: cell proliferation, altered expression of cell cycle regulatory genes in favour of cell proliferation, intracellular reactive oxygen species (ROS) production and mitochondrial depolarization. KV130/131MI may be directly involved in host cell proliferation and hepatocarcinogenesis via altering expression of cell cycle regulatory genes. KV130/131MI may also play pivotal roles in fibrosis and cirrhosis via inducing ROS production and mitochondrial depolarization. Furthermore, these might be the possible reasons for higher occurrence of AG1762/1764TA as compared to A1762T and G1764A in cirrhosis and HCC patients.
Collapse
Affiliation(s)
- Z I Siddiqui
- Hepatitis Research Lab, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - S R Farooqui
- Hepatitis Research Lab, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - S A Azam
- Hepatitis Research Lab, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - M Afroz
- Hepatitis Research Lab, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - S Wajid
- Department of Biotechnology, Jamia Hamdard, New Delhi, India
| | - S Parveen
- Hepatitis Research Lab, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - S N Kazim
- Hepatitis Research Lab, Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
11
|
Austin M, Elliott L, Nicolaou N, Grabowska A, Hulse RP. Breast cancer induced nociceptor aberrant growth and collateral sensory axonal branching. Oncotarget 2017; 8:76606-76621. [PMID: 29100335 PMCID: PMC5652729 DOI: 10.18632/oncotarget.20609] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/15/2017] [Indexed: 12/11/2022] Open
Abstract
The tumour and neuron interaction has a significant impact upon disease progression and the patients quality of life. In breast cancer patients, it is known that there is an interaction between the tumour microenvironment and the sensory neurons to influence the progression of cancer as well as pain, though these mechanisms still need to be clearly defined. Here it is demonstrated that in a rodent orthotopic model of breast cancer (MDA MB 231) there was an increase in nerve fibre innervation into the tumour microenvironment (protein gene product 9.5), which were calcitonin gene related peptide positive C fibre nociceptors. In contrast, there was a reduction in myelinated nerve fibres (NF200). A sensory neuronal cell line was cultured in response to conditioned media from MDA MB231 and MCF7 as well as vascular endothelial growth factor-A (VEGF-A). All these experimental conditions induced sensory neuronal growth, with increased formation of collateral axonal branches. Furthermore, it was demonstrated that MDA MB231 and VEGF-A induced sensory neuronal sensitisation in response to capsaicin a TRPV1 agonist. MDA MB231 induced neuronal growth was suppressed by VEGFR2 inhibition (ZM323881 and neutralising antibody DC101), in addition both MDA MB231 and VEGF-A induced neurite growth was attenuated by the inhibition of ARP2/3 complex through co-treatment with CK666. This demonstrates that breast cancer can interact with the sensory nervous system to drive neuritogenesis through a VEGF-A/VEGFR2/ARP2/3 mediated pathway.
Collapse
Affiliation(s)
- Matt Austin
- Cancer Biology, School of Cancer and Stem Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Laura Elliott
- Cancer Biology, School of Cancer and Stem Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Niovi Nicolaou
- Cancer Biology, School of Cancer and Stem Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Anna Grabowska
- Cancer Biology, School of Cancer and Stem Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Richard P Hulse
- Cancer Biology, School of Cancer and Stem Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
12
|
Magnani L, Patten DK. Fundamental Pathways in Breast Cancer 3: Estrogen Biology. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Liu J, Zhu H, Zhong N, Jiang Z, Xu L, Deng Y, Jiang Z, Wang H, Wang J. Gene silencing of USP1 by lentivirus effectively inhibits proliferation and invasion of human osteosarcoma cells. Int J Oncol 2016; 49:2549-2557. [PMID: 27840911 DOI: 10.3892/ijo.2016.3752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/20/2016] [Indexed: 11/06/2022] Open
Abstract
Osteosarcoma is the most frequent malignant bone tumor, affecting the extremities of adolescents and young adults. Ubiquitin-specific protease 1 (USP1) plays a critical role in many cellular processes including proteasome degradation, chromatin remodeling and cell cycle regulation. In the present study, we discovered that USP1 was overexpressed in 26 out of 30 osteosarcoma tissues compared to cartilage tumor tissues and normal bone tissues. We then constructed a lentiviral vector mediating RNA interference (RNAi) targeting USP1 and demonstrated that it significantly suppressed the mRNA and protein expression of the USP1 gene in U2OS cells. Knockdown of USP1 inhibited the growth and colony-forming, as well as significantly reduced the invasiveness of U2OS cells. Western blot analysis indicated that suppression of USP1 downregulated the expression of many proteins including SIK2, MMP-2, GSK-3β, Bcl-2, Stat3, cyclin E1, Notch1, Wnt-1 and cyclin A1. Most of these proteins are associated with tumor genesis and development. RNAi of SIK2 significantly decreased SIK2 protein expression and inhibited the ability of forming colonies, as well as induced apoptosis and reduced the invasiveness of U2OS cells. Collectively, our results suggest that silencing USP1 inhibits cell proliferation and invasion in U2OS cells. Therefore, USP1 may provide a novel therapeutic target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Jinbo Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Hongjun Zhu
- Department of Thoracic Surgery, The First People's Hospital of Shangqiu, Shangqiu, Henan 476100, P.R. China
| | - Ning Zhong
- Department of Thoracic Surgery, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, Jiangsu 215000, P.R. China
| | - Zifeng Jiang
- Clinical Laboratories, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lele Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| | - Youping Deng
- Department of Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Zhenhuan Jiang
- Department of Orthopaedics, People's Hospital of Yixing City, Yixing, Jiangsu 214200, P.R. China
| | - Hongwei Wang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jinzhi Wang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
14
|
Yan F, Wang X, Zhu M, Hu X. RNAi-mediated downregulation of cyclin Y to attenuate human breast cancer cell growth. Oncol Rep 2016; 36:2793-2799. [PMID: 27666310 DOI: 10.3892/or.2016.5126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/26/2015] [Indexed: 11/06/2022] Open
Abstract
Cyclin Y (CCNY) is a newly identified PFTK1 interacting protein and has been found to be associated with the proliferation and tumorigenesis of human non-small cell lung cancer. In the present study, we analyzed the expression levels of CCNY in 65 cases of breast cancer (BC) tissues and in four BC cell lines, BT-474, MDA-MB-231, T-47D and MCF-7. Lentivirus-mediated short hairpin RNA (shRNA) was employed to knock down CCNY expression in MCF-7 and MDA-MB-231 cells. The effects of CCNY depletion on cell growth were examined by MTT, colony formation and flow cytometry assays. The results showed that immunohistochemical expression of CCNY in tumor tissues is stronger than that in normal tissues. CCNY was also expressed in all four BC cells. The knockdown of CCNY resulted in a significant reduction in cell proliferation and colony formation ability. Cell cycle analysis showed that CCNY knockdown arrested MDA-MB‑231 cells in the G0/G1 phase. Furthermore, depletion of CCNY inhibited BC cell growth via the activation of Bad and GSK3β, as well as cleavages of PARP and caspase-3 in a p53-dependent manner. Therefore, we believe that CCNY has biological effect in BC development, and its inhibition via an RNA interference lentiviral system may provide a therapeutic option for BC.
Collapse
Affiliation(s)
- Feng Yan
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, P.R. China
| | - Xiaoming Wang
- Department of Clinical Laboratory, Nanjing Medical University Cancer Hospital and Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Mingchen Zhu
- Department of Clinical Laboratory, Nanjing Medical University Cancer Hospital and Jiangsu Cancer Hospital, Nanjing, Jiangsu 210009, P.R. China
| | - Xiaoya Hu
- College of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, P.R. China
| |
Collapse
|
15
|
Miftakhova R, Hedblom A, Batkiewicz L, Anagnosaki L, Zhang Y, Sjölander A, Wingren AG, Wolgemuth DJ, Persson JL. Cyclin A1 regulates the interactions between mouse haematopoietic stem and progenitor cells and their niches. Cell Cycle 2016; 14:1948-60. [PMID: 25785996 DOI: 10.1080/15384101.2015.1026513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
It remains poorly understood how the haematopoietic stem/progenitor cells (HSPC) are attracted to their niches and the functional consequences of such interaction. In the present study, we show that the cell cycle regulator cyclin A1 in association with vascular endothelial growth factor receptor 1 (VEGFR1), is required for HSPC and their niches to maintain their function and proper interaction. In the absence of cyclin A1, the HSPC in the BM are increased in their frequency and display an increased migratory and homing ability. Concomitantly, the ability of the endosteal and central BM niche zones to attract and home the wild-type HSPC is significantly reduced in cyclin A1-null mice as compared to the wild-type controls. The impaired proliferation and homing of HSPC in the BM of cyclin A1-null mice are attributed to the increased density of microvessels in the endosteal and central BM niche zones, which is associated with the increased VEGFR1 expression. Thus, modulation of cyclin A1 and VEGFR1 in HSPC and their niches may provide new insights into therapeutic approaches.
Collapse
Affiliation(s)
- Regina Miftakhova
- a Division of Experimental Cancer Research; Department of Translational Medicine; Clinical Research Center; Lund University ; Malmö , Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Proteomic Analysis of Stage-II Breast Cancer from Formalin-Fixed Paraffin-Embedded Tissues. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3071013. [PMID: 27110560 PMCID: PMC4823502 DOI: 10.1155/2016/3071013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/22/2016] [Accepted: 03/07/2016] [Indexed: 01/25/2023]
Abstract
Breast cancer is the most frequently occurring disease among women worldwide. The early stage of breast cancer identification is the key challenge in cancer control and prevention procedures. Although gene expression profiling helps to understand the molecular mechanism of diseases or disorder in the living system, gene expression pattern alone is not sufficient to predict the exact mechanisms. Current proteomics tools hold great application for analysis of cancerous conditions. Hence, the generation of differential protein expression profiles has been optimized for breast cancer and normal tissue samples in our organization. Normal and tumor tissues were collected from 20 people from a local hospital. Proteins from the diseased and normal tissues have been investigated by 2D gel electrophoresis and MALDI-TOF-MS. The peptide mass fingerprint data were fed into various public domains like Mascot, MS-Fit, and Pept-ident against Swiss-Prot protein database and the proteins of interest were identified. Some of the differentially expressed proteins identified were human annexin, glutathione S-transferase, vimentin, enolase-1, dihydrolipoamide dehydrogenase, glutamate dehydrogenase, Cyclin A1, hormone sensitive lipase, beta catenin, and so forth. Many types of proteins were identified as fundamental steps for developing molecular markers for diagnosis of human breast cancer as well as making a new proteomic database for future research.
Collapse
|
17
|
Miftakhova R, Hedblom A, Semenas J, Robinson B, Simoulis A, Malm J, Rizvanov A, Heery DM, Mongan NP, Maitland NJ, Allegrucci C, Persson JL. Cyclin A1 and P450 Aromatase Promote Metastatic Homing and Growth of Stem-like Prostate Cancer Cells in the Bone Marrow. Cancer Res 2016; 76:2453-64. [PMID: 26921336 DOI: 10.1158/0008-5472.can-15-2340] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/17/2016] [Indexed: 11/16/2022]
Abstract
Bone metastasis is a leading cause of morbidity and mortality in prostate cancer. While cancer stem-like cells have been implicated as a cell of origin for prostate cancer metastasis, the pathways that enable metastatic development at distal sites remain largely unknown. In this study, we illuminate pathways relevant to bone metastasis in this disease. We observed that cyclin A1 (CCNA1) protein expression was relatively higher in prostate cancer metastatic lesions in lymph node, lung, and bone/bone marrow. In both primary and metastatic tissues, cyclin A1 expression was also correlated with aromatase (CYP19A1), a key enzyme that directly regulates the local balance of androgens to estrogens. Cyclin A1 overexpression in the stem-like ALDH(high) subpopulation of PC3M cells, one model of prostate cancer, enabled bone marrow integration and metastatic growth. Further, cells obtained from bone marrow metastatic lesions displayed self-renewal capability in colony-forming assays. In the bone marrow, cyclin A1 and aromatase enhanced local bone marrow-releasing factors, including androgen receptor, estrogen and matrix metalloproteinase MMP9 and promoted the metastatic growth of prostate cancer cells. Moreover, ALDH(high) tumor cells expressing elevated levels of aromatase stimulated tumor/host estrogen production and acquired a growth advantage in the presence of host bone marrow cells. Overall, these findings suggest that local production of steroids and MMPs in the bone marrow may provide a suitable microenvironment for ALDH(high) prostate cancer cells to establish metastatic growths, offering new approaches to therapeutically target bone metastases. Cancer Res; 76(8); 2453-64. ©2016 AACR.
Collapse
Affiliation(s)
- Regina Miftakhova
- Department of Translational Medicine, Lund University, Lund, Sweden. Department of Genetics, Kazan Federal University, Tatarstan, Russia
| | - Andreas Hedblom
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Julius Semenas
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Brian Robinson
- Department of Pathology, Weill Cornell Medical College, New York, New York
| | - Athanasios Simoulis
- Department of Clinical Pathology and Cytology, Skåne University Hospital, Scania, Sweden
| | - Johan Malm
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Albert Rizvanov
- Department of Genetics, Kazan Federal University, Tatarstan, Russia
| | - David M Heery
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Nigel P Mongan
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Norman J Maitland
- Yorkshire Cancer Research Unit, University of York, York, United Kingdom
| | - Cinzia Allegrucci
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jenny L Persson
- Department of Translational Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
18
|
Wang C, Luo Y, Lu J, Wang Y, Sheng G. Polydatin Induces Apoptosis and Inhibits Growth of Acute Monocytic Leukemia Cells. J Biochem Mol Toxicol 2015; 30:200-5. [PMID: 26616494 DOI: 10.1002/jbt.21779] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/11/2015] [Accepted: 10/23/2015] [Indexed: 01/30/2023]
Abstract
Polydatin (PD), a component isolated from Polygonum cuspidatum, has various activities such as inhibiting platelet aggregation, lowering level of blood lipid, reducing lipid peroxidation, and so on. However, the antitumor activity of PD has been poorly reported. In the present study, effect of PD on cell proliferation was evaluated by Cell Counting Kit-8, and cell cycle and apoptosis were investigated by flow cytometry. Meanwhile, the protein expression level of Bc1-2, Bax, cyclin A, cyclin B, and cyclin D1, which associated with apoptosis and cell cycle were analyzed by Western blotting. Results show that PD could effectively inhibit the growth, arrest cells in S phase, and induce apoptosis of acute monocytic leukemia cell line THP-1; meanwhile, expression of cyclin D1 and Bc1-2 decreased significantly, and expression of Bax and cyclin A increased notably. All results suggest that PD maybe a potential therapeutic strategy for acute monocytic leukemia.
Collapse
Affiliation(s)
- Chunmei Wang
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China.
| | - Yuan Luo
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Jie Lu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Yingchao Wang
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| | - Guangyao Sheng
- Department of Pediatrics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, People's Republic of China
| |
Collapse
|
19
|
Bäumer N, Bäumer S, Haak M, Koschmieder S, Schönig K, Berdel WE, Müller-Tidow C. A Limited Role for the Cell Cycle Regulator Cyclin A1 in Murine Leukemogenesis. PLoS One 2015; 10:e0129147. [PMID: 26080083 PMCID: PMC4469679 DOI: 10.1371/journal.pone.0129147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/05/2015] [Indexed: 12/04/2022] Open
Abstract
The quest for novel therapeutic targets in acute myeloid leukemia (AML) is still ongoing. One of such targets, cyclin A1, was shown to be overexpressed in AML including AML stem cells. However, the function of cyclin A1 in AML is largely unknown, and the data on its impact on patients´ survival remain controversial. Therefore, we developed a transgenic mouse model of stem cell-directed inducible cyclin A1 overexpression and crossed these mice with PML-RARα-knockin mice, which develop an AML M3-like phenotype. To observe the effects of cyclin A1 loss-of-function, we also crossed PML-RARα-knockin mice to cyclin A1-knockout mice. Neither overexpression nor loss of cyclin A1 significantly altered leukemogenesis in PML-RARα-knockin mice. These findings imply that upregulation of cyclin A1 is not essential for leukemogenesis. Our data suggest that cyclin A1 does not represent a suitable target for AML therapy.
Collapse
Affiliation(s)
- Nicole Bäumer
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- * E-mail: (NB); (CMT)
| | - Sebastian Bäumer
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Miriam Haak
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Steffen Koschmieder
- Department of Medicine (Hematology, Oncology, Hemostaseology, and SCT), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Kai Schönig
- Central Institute of Mental Health, Department of Molecular Biology, Heidelberg University, Mannheim, Germany
| | - Wolfgang E. Berdel
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- Dept. of Medicine IV, Hematology and Oncology, University of Halle, Halle, Germany
- * E-mail: (NB); (CMT)
| |
Collapse
|
20
|
Correlation of CCNA1 promoter methylation with malignant tumors: a meta-analysis introduction. BIOMED RESEARCH INTERNATIONAL 2015; 2015:134027. [PMID: 25654082 PMCID: PMC4310450 DOI: 10.1155/2015/134027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 10/23/2014] [Accepted: 11/07/2014] [Indexed: 01/09/2023]
Abstract
Epigenetic silencing of tumor suppressor genes by promoter methylation plays vital roles in the process of carcinogenesis. The purpose of this meta-analysis was to determine whether the aberrant methylation of cyclin A1 (CCNA1) may be of great significance to human malignant tumors. By searching both English and Chinese language-based electronic databases carefully, we tabulated and analyzed parameters from each study. All human-associated case-control studies were included providing available data for CCNA1 methylation and reporting the adjusted odds ratios (ORs) and 95% confidence intervals (CI) conducted with the use of Version 12.0 STATA software. A total of 10 case-control studies (619 patients with cancers and 292 healthy controls) were included for the following statistical analysis. Pooled OR values from all articles revealed that the frequency of CCNA1 methylation in cancer tissues was significantly higher than those of normal tissues (P < 0.001). Further ethnicity indicated that the frequency of CCNA1 methylation was correlated with the development of malignant tumors among all those included experimental subgroups (all P < 0.05). These data from results indicated a significant connection of CCNA1 methylation with poor progression in human malignant tumors among both Caucasian and Asian populations.
Collapse
|
21
|
Liu Q, Su PF, Zhao S, Shyr Y. Transcriptome-wide signatures of tumor stage in kidney renal clear cell carcinoma: connecting copy number variation, methylation and transcription factor activity. Genome Med 2014; 6:117. [PMID: 25648588 PMCID: PMC4293006 DOI: 10.1186/s13073-014-0117-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 11/26/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Comparative analysis of expression profiles between early and late stage cancers can help to understand cancer progression and metastasis mechanisms and to predict the clinical aggressiveness of cancer. The observed stage-dependent expression changes can be explained by genetic and epigenetic alterations as well as transcription dysregulation. Unlike genetic and epigenetic alterations, however, activity changes of transcription factors, generally occurring at the post-transcriptional or post-translational level, are hard to detect and quantify. METHODS Here we developed a statistical framework to infer the activity changes of transcription factors by simultaneously taking into account the contributions of genetic and epigenetic alterations to mRNA expression variations. RESULTS Applied to kidney renal clear cell carcinoma (KIRC), the model underscored the role of methylation as a significant contributor to stage-dependent expression alterations and identified key transcription factors as potential drivers of cancer progression. CONCLUSIONS Integrating copy number, methylation, and transcription factor activity signatures to explain stage-dependent expression alterations presented a precise and comprehensive view on the underlying mechanisms during KIRC progression.
Collapse
Affiliation(s)
- Qi Liu
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Shilin Zhao
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240 China
| |
Collapse
|
22
|
Interleukin-17 receptor expression on vascular endothelial cells of masses of skeletal extramedullary disease in myeloma patients. Pathol Res Pract 2014; 210:586-90. [DOI: 10.1016/j.prp.2014.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/13/2014] [Accepted: 04/30/2014] [Indexed: 12/12/2022]
|